1
|
Xie Y, Mi X, Xing Y, Dai Z, Pu Q. Past, present, and future of exosomes research in cancer: A bibliometric and visualization analysis. Hum Vaccin Immunother 2025; 21:2488551. [PMID: 40207548 PMCID: PMC11988232 DOI: 10.1080/21645515.2025.2488551] [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: 01/21/2025] [Revised: 03/12/2025] [Accepted: 04/01/2025] [Indexed: 04/11/2025] Open
Abstract
Cancer seriously threatens the lives and health of people worldwide, and exosomes seem to play an important role in managing cancer effectively, which has attracted extensive attention from researchers in recent years. This study aimed to scientifically visualize exosomes research in cancer (ERC) through bibliometric analysis, reviewing the past, summarizing the present, and predicting the future, with a view to providing valuable insights for scholars and policy makers. Researches search and data collection from Web of Science Core Collection and clinical trial.gov. Calculations and visualizations were performed using Microsoft Excel, VOSviewer, Bibliometrix R-package, and CiteSpace. As of December 1, 2024, and March 8, 2025, we identified 8,001 ERC-related publications and 107 ERC-related clinical trials, with an increasing trend in annual publications. Our findings supported that China, Nanjing Medical University, and International Journal of Molecular Sciences were the most productive countries, institutions, and journals, respectively. Whiteside, Theresa L. had the most publications, while Théry, C was the most co-cited scholar. In addition, Cancer Research was the most co-cited journal. Spatial and temporal distribution of clinical trials was the same as for publications. High-frequency keywords were "extracellular vesicle," "microRNA" and "biomarker." Additional, "surface functionalization," "plant," "machine learning," "nanomaterials," "promotes metastasis," "engineered exosomes," and "macrophage-derived exosomes" were promising research topics. Our study comprehensively and visually summarized the structure, hotspots, and evolutionary trends of ERC. It would inspire subsequent studies from a macroscopic perspective and provide a basis for rational allocation of resources and identification of collaborations among researchers.
Collapse
Affiliation(s)
- Yafei Xie
- Department of Thoracic Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Xingqi Mi
- Department of Thoracic Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Yikai Xing
- Department of Thoracic Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Zhangyi Dai
- Department of Thoracic Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Qiang Pu
- Department of Thoracic Surgery, West China Hospital of Sichuan University, Chengdu, China
| |
Collapse
|
2
|
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
|
3
|
Chen T, Ma W, Wang X, Ye Q, Hou X, Wang Y, Jiang C, Meng X, Sun Y, Cai J. Insights of immune cell heterogeneity, tumor-initiated subtype transformation, drug resistance, treatment and detecting technologies in glioma microenvironment. J Adv Res 2025; 72:527-554. [PMID: 39097088 DOI: 10.1016/j.jare.2024.07.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Revised: 06/30/2024] [Accepted: 07/29/2024] [Indexed: 08/05/2024] Open
Abstract
BACKGROUND With the gradual understanding of glioma development and the immune microenvironment, many immune cells have been discovered. Despite the growing comprehension of immune cell functions and the clinical application of immunotherapy, the precise roles and characteristics of immune cell subtypes, how glioma induces subtype transformation of immune cells and its impact on glioma progression have yet to be understood. AIM OF THE REVIEW In this review, we comprehensively center on the four major immune cells within the glioma microenvironment, particularly neutrophils, macrophages, lymphocytes, myeloid-derived suppressor cells (MDSCs), and other significant immune cells. We discuss (1) immune cell subtype markers, (2) glioma-induced immune cell subtype transformation, (3) the mechanisms of each subtype influencing chemotherapy resistance, (4) therapies targeting immune cells, and (5) immune cell-associated single-cell sequencing. Eventually, we identified the characteristics of immune cell subtypes in glioma, comprehensively summarized the exact mechanism of glioma-induced immune cell subtype transformation, and concluded the progress of single-cell sequencing in exploring immune cell subtypes in glioma. KEY SCIENTIFIC CONCEPTS OF REVIEW In conclusion, we have analyzed the mechanism of chemotherapy resistance detailly, and have discovered prospective immunotherapy targets, excavating the potential of novel immunotherapies approach that synergistically combines radiotherapy, chemotherapy, and surgery, thereby paving the way for improved immunotherapeutic strategies against glioma and enhanced patient outcomes.
Collapse
Affiliation(s)
- Tongzheng Chen
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Wenbin Ma
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xin Wang
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Qile Ye
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xintong Hou
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yiwei Wang
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Chuanlu Jiang
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China; The Six Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xiangqi Meng
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.
| | - Ying Sun
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.
| | - Jinquan Cai
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.
| |
Collapse
|
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
|
Bandyopadhyay A, Sinha S, Roy R, Biswas N. Autophagy mediated immune response regulation and drug resistance in cancer. Mol Biol Rep 2025; 52:492. [PMID: 40402380 DOI: 10.1007/s11033-025-10573-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Accepted: 05/02/2025] [Indexed: 05/23/2025]
Abstract
Autophagy is a critical regulator of cellular homeostasis. The proteins involved in autophagy orchestrate the functions to strike the balance between cell survival and cell death in context-specific situations like aging, infections, inflammation and most importantly carcinogenesis. One of the major dead-locks in cancer treatment is the development of resistance to the available drugs (multi-drug resistance) as well as immune-suppressions in patients. Different studies over time have shown that autophagy is being involved in chemotherapy by working hand in hand with apoptosis or drug resistance through proliferative signals. Resistance to various drugs, such as, Cisplatin, Vincristine, Tamoxifen (TAM) occurs by epigenetic modifications, changed expression levels of microRNAs (miRNAs/miRs), and long non-coding RNAs (lncRNAs), which are regulated by the aberrant autophagy levels in lung, and breast cancers. More interestingly in the tumour microenvironment the immune suppressor cells also bring in autophagy in different pathway regulations either helping or opposing the whole carcinogenesis process. Macrophages, T cells, B cells, dendritic cells (DCs), neutrophils, and fibroblasts show involvement of autophagy in their differentiation and development in the tumor microenvironment (TME). Here, this extensive review for the first time tries to bring under a single canopy, several recent examples of autophagy-mediated immune regulations and autophagy-mediated epigenetically regulated drug resistance in different types of cancers.
Collapse
Affiliation(s)
- Anupriya Bandyopadhyay
- Department of Life Sciences, Presidency University, 86/1 College Street, Kolkata, 700073, India
| | - Samraj Sinha
- Department of Life Sciences, Presidency University, 86/1 College Street, Kolkata, 700073, India
| | - Rajdeep Roy
- Department of Life Sciences, Presidency University, 86/1 College Street, Kolkata, 700073, India
| | - Nabendu Biswas
- Department of Life Sciences, Presidency University, 86/1 College Street, Kolkata, 700073, India.
| |
Collapse
|
6
|
Zhao W, Zhang Z, Xie M, Ding F, Zheng X, Sun S, Du J. Exploring tumor-associated macrophages in glioblastoma: from diversity to therapy. NPJ Precis Oncol 2025; 9:126. [PMID: 40316746 PMCID: PMC12048723 DOI: 10.1038/s41698-025-00920-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Accepted: 04/22/2025] [Indexed: 05/04/2025] Open
Abstract
Glioblastoma is the most aggressive and lethal cancer of the central nervous system, presenting substantial treatment challenges. The current standard treatment, which includes surgical resection followed by temozolomide and radiation, offers limited success. While immunotherapies, such as immune checkpoint inhibitors, have proven effective in other cancers, they have not demonstrated significant efficacy in GBM. Emerging research highlights the pivotal role of tumor-associated macrophages (TAMs) in supporting tumor growth, fostering treatment resistance, and shaping an immunosuppressive microenvironment. Preclinical studies show promising results for therapies targeting TAMs, suggesting potential in overcoming these barriers. TAMs consist of brain-resident microglia and bone marrow-derived macrophages, both exhibiting diverse phenotypes and functions within the tumor microenvironment. This review delves into the origin, heterogeneity, and functional roles of TAMs in GBM, underscoring their dual roles in tumor promotion and suppression. It also summarizes recent progress in TAM-targeted therapies, which may, in combination with other treatments like immunotherapy, pave the way for more effective and personalized strategies against this aggressive malignancy.
Collapse
Affiliation(s)
- Wenwen Zhao
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Zhi Zhang
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Mingyuan Xie
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Feng Ding
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Xiangrong Zheng
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Shicheng Sun
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Jianyang Du
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China.
| |
Collapse
|
7
|
Zhang L, Wang K, Li F, Zhang L, Wu L, Tie R, Litifu K, Fu Y, Liu S, Ni J, Chang P, Xu J, Zhao H, Liu L. Ribosomal protein S3A (RPS3A), as a transcription regulator of colony-stimulating factor 1 (CSF1), promotes glioma progression through regulating the recruitment and autophagy-mediated M2 polarization of tumor-associated macrophages. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:5437-5452. [PMID: 39560749 DOI: 10.1007/s00210-024-03601-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 11/01/2024] [Indexed: 11/20/2024]
Abstract
Dysregulated expression of ribosomal protein S3A (RPS3A) is associated with the tissue infiltration of immune-related cells in a variety of cancers. However, the role of RPS3A in immune cell infiltration in glioma remains unclear. This study aimed to explore the role of RPS3A in the glioma immune microenvironment. RPS3A expression was detected in tumor tissues from patients with glioma. U251 cells were transfected with RPS3A shRNA (sh-RPS3A) and overexpression vector (pcDNA-RPS3A) and then co-cultured with PMA-induced THP-1 cells. Cell viability, invasion, and apoptosis were detected by Edu staining, Transwell, and flow cytometry, respectively. The expression of tumor-associated macrophage (TAM) M1 and M2 markers was detected with RT-qPCR. Next, the interaction between RPS3A and E4 transcription factor 1 (E4F1) was verified by Co-IP analysis, and the binding of E4F1 to colony-stimulating factor 1 (CSF1) promoter was verified by ChIP analysis. Overexpression vectors of CSF1 and E4F1 were used to treat sh-RPS3A-transfected U251 cells for reversal experiments. Finally, U251 cells transfected with sh-RPS3A adenovirus vectors were subcutaneously injected into nude mice to construct a xenograft tumor model, and the growth and metastasis of glioma in vivo were monitored. RPS3A was significantly upregulated in glioma tissues. Overexpression of RPS3A promoted glioma cell proliferation and invasion and inhibited apoptosis. Moreover, overexpression of RPS3A promoted TAM proliferation, invasion, and M2 polarization. Silencing RPS3A had the opposite effect. Silencing RPS3A inhibited autophagy in U251 cells, whereas rapamycin, an activator of autophagy, reversed the inhibitory effect of RPS3A silencing on TAM M2 polarization. Meanwhile, RPS3A promoted its expression by interacting with E4F1, and E4F1 promoted CSF1 transcriptional activation. Overexpression of CSF1 promoted the proliferation and invasion of U251 cells and reversed the inhibitory effect of RPS3A silencing on TAM proliferation and invasion, but had no effect on TAM M2 polarization. The results of in vivo experiments showed that knockdown of RPS3A significantly inhibited glioma tumor growth and metastasis in vivo. This study revealed that RPS3A recruited TAMs by upregulating E4F1-mediated transcription activation of CSF1, and promoted the M2 polarization of TAMs through autophagy, promoting glioma cell malignant growth and tumor progression.
Collapse
Affiliation(s)
- Liang Zhang
- Northwest University, Guodu Education and Technology Industrial Zone, No. 1 Xuefu Street, Chang'an District, Xi'an, 710127, China
- Xi'an Daxing Hospital, No. 353 Laodong North Road, Lianhu District, Xi'an, 710016, China
- Second Affiliated Hospital of Xi'an Medical University, No. 167 Fangdong Street, Xi'an, 710038, China
| | - Kun Wang
- Second Affiliated Hospital of Xi'an Medical University, No. 167 Fangdong Street, Xi'an, 710038, China
| | - Fei Li
- Second Affiliated Hospital of Xi'an Medical University, No. 167 Fangdong Street, Xi'an, 710038, China
| | - Lingxue Zhang
- Second Affiliated Hospital of Xi'an Medical University, No. 167 Fangdong Street, Xi'an, 710038, China
| | - Lin Wu
- Second Affiliated Hospital of Xi'an Medical University, No. 167 Fangdong Street, Xi'an, 710038, China
| | - Ru Tie
- Second Affiliated Hospital of Xi'an Medical University, No. 167 Fangdong Street, Xi'an, 710038, China
| | - Kamulan Litifu
- Second Affiliated Hospital of Xi'an Medical University, No. 167 Fangdong Street, Xi'an, 710038, China
| | - Yujie Fu
- Second Affiliated Hospital of Xi'an Medical University, No. 167 Fangdong Street, Xi'an, 710038, China
| | - Simeng Liu
- Second Affiliated Hospital of Xi'an Medical University, No. 167 Fangdong Street, Xi'an, 710038, China
| | - Jiaxin Ni
- Second Affiliated Hospital of Xi'an Medical University, No. 167 Fangdong Street, Xi'an, 710038, China
| | - Pan Chang
- Second Affiliated Hospital of Xi'an Medical University, No. 167 Fangdong Street, Xi'an, 710038, China
| | - Jun Xu
- Xi'an Daxing Hospital, No. 353 Laodong North Road, Lianhu District, Xi'an, 710016, China
| | - Haikang Zhao
- Second Affiliated Hospital of Xi'an Medical University, No. 167 Fangdong Street, Xi'an, 710038, China.
| | - Lingtong Liu
- Department of Neurosurgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, No.32 West Second Section First Ring Road, Chengdu, 610072, China.
| |
Collapse
|
8
|
Ramezani A, Rahnama M, Mahmoudian F, Shirazi F, Ganji M, Bakhshi S, Khalesi B, Hashemi ZS, Khalili S. Current Understanding of the Exosomes and Their Associated Biomolecules in the Glioblastoma Biology, Clinical Treatment, and Diagnosis. J Neuroimmune Pharmacol 2025; 20:48. [PMID: 40299204 DOI: 10.1007/s11481-025-10204-7] [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: 07/23/2024] [Accepted: 04/08/2025] [Indexed: 04/30/2025]
Abstract
Glioblastoma is the most common and aggressive brain tumor with a low survival rate. Due to its heterogeneous composition, high invasiveness, and frequent recurrence after surgery, treatment success has been limited. In addition, due to the brain's unique immune status and the suppressor tumor microenvironment (TME), glioblastoma treatment has faced more challenges. Exosomes play a critical role in cancer metastasis by regulating cell-cell interactions that promote tumor growth, angiogenesis, metastasis, treatment resistance, and immunological regulation in the tumor microenvironment. This review explores the pivotal role of exosomes in the development of glioblastoma, with a focus on their potential as non-invasive biomarkers for prognosis, early detection and real-time monitoring of disease progression. Notably, exosome-based drug delivery methods hold promise for overcoming the blood-brain barrier (BBB) and developing targeted therapies for glioblastoma. Despite challenges in clinical translation, the potential for personalized exosome = -054321`therapies and the capacity to enhance therapeutic responses in glioblastoma, present intriguing opportunities for improving patient outcomes. It seems that getting a good and current grasp of the role of exosomes in the fight against glioblastoma would properly serve the scientific community to further their understanding of the related potentials of these biological moieties.
Collapse
Affiliation(s)
- Aghdas Ramezani
- Department of Molecular Imaging, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Maryam Rahnama
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Fatemeh Mahmoudian
- Cancer Research Center, Semnan University of Medical Sciences, Semnan, Iran
| | - Fatemeh Shirazi
- Division of Genetics, Department of Cell and Molecular Biology and Microbiology, Faculty of Biological Sciences and Technologies, University of Isfahan, Isfahan, Iran
| | - Mahmoud Ganji
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Shohreh Bakhshi
- Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Bahman Khalesi
- Department of Research and Production of Poultry Viral Vaccine, Education and Extension Organization, Razi Vaccine and Serum Research Institute, Agricultural Research, Karaj, 3197619751, Iran
| | - Zahra Sadat Hashemi
- ATMP Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran.
| | - Saeed Khalili
- Department of Biology Sciences, Shahid Rajaee Teacher Training University, Tehran, Iran.
| |
Collapse
|
9
|
Fuse H, Zheng Y, Alzoubi I, Graeber MB. TAMing Gliomas: Unraveling the Roles of Iba1 and CD163 in Glioblastoma. Cancers (Basel) 2025; 17:1457. [PMID: 40361384 PMCID: PMC12070867 DOI: 10.3390/cancers17091457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2025] [Revised: 04/19/2025] [Accepted: 04/23/2025] [Indexed: 05/15/2025] Open
Abstract
Gliomas, the most common type of primary brain tumor, are a significant cause of morbidity and mortality worldwide. Glioblastoma, a highly malignant subtype, is particularly common, aggressive, and resistant to treatment. The tumor microenvironment (TME) of gliomas, especially glioblastomas, is characterized by a distinct presence of tumor-associated macrophages (TAMs), which densely infiltrate glioblastomas, a hallmark of these tumors. This macrophage population comprises both tissue-resident microglia as well as macrophages derived from the walls of blood vessels and the blood stream. Ionized calcium-binding adapter molecule 1 (Iba1) and CD163 are established cellular markers that enable the identification and functional characterization of these cells within the TME. This review provides an in-depth examination of the roles of Iba1 and CD163 in malignant gliomas, with a focus on TAM activation, migration, and immunomodulatory functions. Additionally, we will discuss how recent advances in AI-enhanced cell identification and visualization techniques have begun to transform the analysis of TAMs, promising unprecedented precision in their characterization and providing new insights into their roles within the TME. Iba1 and CD163 appear to have both unique and shared roles in glioma pathobiology, and both have the potential to be targeted through different molecular and cellular mechanisms. We discuss the therapeutic potential of Iba1 and CD163 based on available preclinical (experimental) and clinical (human tissue-based) evidence.
Collapse
Affiliation(s)
- Haneya Fuse
- School of Medicine, Sydney Campus, University of Notre Dame, 160 Oxford Street, Sydney, NSW 2010, Australia;
| | - Yuqi Zheng
- Ken Parker Brain Tumor Research Laboratories, Brain and Mind Centre, Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2050, Australia;
| | - Islam Alzoubi
- School of Computer Science, The University of Sydney, J12/1 Cleveland St, Sydney, NSW 2008, Australia;
| | - Manuel B. Graeber
- Ken Parker Brain Tumor Research Laboratories, Brain and Mind Centre, Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2050, Australia;
- University of Sydney Association of Professors (USAP), University of Sydney, Sydney, NSW 2006, Australia
| |
Collapse
|
10
|
Tang Z, Xue Z, Liu X, Zhang Y, Zhao J, Liu J, Zhang L, Guo Q, Feng B, Wang J, Zhang D, Li X. Inhibition of hypoxic exosomal miR-423-3p decreases glioma progression by restricting autophagy in astrocytes. Cell Death Dis 2025; 16:265. [PMID: 40199864 PMCID: PMC11978802 DOI: 10.1038/s41419-025-07576-2] [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: 07/16/2024] [Revised: 02/23/2025] [Accepted: 03/18/2025] [Indexed: 04/10/2025]
Abstract
The tumor microenvironment (TME) of gliomas comprises glioma cells and surrounding cells, such as astrocytes, macrophages, T cells, and neurons. In the TME, glioma cells can activate normal human astrocytes (NHAs) through the secretion of exosomes and the activation of astrocytes can further improve the progression of glioma, leading to a poor prognosis for patients. However, the molecular mechanisms underlying NHAs activation by gliomas remain largely unknown. It this study, glioma-derived exosomes (GDEs) play an important role in the modulation of autophagy and activation of NHAs. Compared with normoxic GDEs, hypoxic glioma-derived exosomes (H-GDEs) further improved autophagy and activation of astrocytes, which strongly promoted the progression of glioma cells. In an miRNA array between two types of exosomes from gliomas, miR-423-3p was highly expressed in H-GDEs and played an important role in autophagy, resulting in the activation of NHAs. The mechanism by which hypoxic glioma cells react with NHAs to create an immunosuppressive microenvironment was identified and 15d-PGJ2 was established as an effective inhibitor of miR-423-3p to suppress NHAs activation. These findings provide new insights into the diagnosis and treatment of gliomas by targeting autophagy and miR-423-3p expression.
Collapse
Affiliation(s)
- Ziyi Tang
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
- Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Health and Function Remodeling, Jinan, China
| | - Zhiwei Xue
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
- Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Health and Function Remodeling, Jinan, China
| | - Xuchen Liu
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
- Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Health and Function Remodeling, Jinan, China
| | - Yan Zhang
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
- Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Health and Function Remodeling, Jinan, China
| | - Jiangli Zhao
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
- Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Health and Function Remodeling, Jinan, China
| | - Junzhi Liu
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
- Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Health and Function Remodeling, Jinan, China
| | - Lin Zhang
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Qindong Guo
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
- Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Health and Function Remodeling, Jinan, China
| | - Bowen Feng
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
- Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Health and Function Remodeling, Jinan, China
| | - Jiwei Wang
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China.
- Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Health and Function Remodeling, Jinan, China.
| | - Di Zhang
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China.
- Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Health and Function Remodeling, Jinan, China.
| | - Xingang Li
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China.
- Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Health and Function Remodeling, Jinan, China.
| |
Collapse
|
11
|
Niazi V, Ghafouri-Fard S. Effect of hypoxia on extracellular vesicles in malignant and non-malignant conditions. Cancer Treat Res Commun 2025; 43:100924. [PMID: 40209539 DOI: 10.1016/j.ctarc.2025.100924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2025] [Revised: 03/21/2025] [Accepted: 04/04/2025] [Indexed: 04/12/2025]
Abstract
Extracellular vesicles (EVs) are produced by virtually all types of cells and can be detected in nearly all extracellular places. These particles mediate intercellular communication and transfer their cargo to the recipient cells, inducing a variety of processes in these cells through transmission of several biomolecules such as miRNAs, lncRNAs, other transcripts and a variety of proteins. It has been documented that size, quantity, and expression of biomolecules in the EVs are influenced by the level of oxygen. In fact, hypoxia can affect several cellular processes through modulation of the cargo of these vesicles. Hypoxic exosomes derived from tumor cells have several protumoral effects on the recipient cells, including enhancement of proliferation, migration, and invasion in other tumoral cells, induction of metastasis in distant organs, stimulation of angiogenesis in the endothelial cells, and modulation of macrophage polarization. Hypoxic EVs also contribute to several non-malignant diseases. This review summarizes the effect of hypoxia on EVs cargo in malignant and nonmalignant diseases of different organs.
Collapse
Affiliation(s)
- Vahid Niazi
- Stem Cell Research Center, Golestan University of Medical Science, Gorgan, Iran; School of Advanced Technologies in Medicine, Golestan University of Medical Science, Gorgan, Iran
| | - Soudeh Ghafouri-Fard
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
12
|
Gao Y, Zhang M, Wang G, Lai W, Liao S, Chen Y, Ning Q, Tang S. Metabolic cross-talk between glioblastoma and glioblastoma-associated microglia/macrophages: From basic insights to therapeutic strategies. Crit Rev Oncol Hematol 2025; 208:104649. [PMID: 39922398 DOI: 10.1016/j.critrevonc.2025.104649] [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: 01/06/2025] [Revised: 01/26/2025] [Accepted: 02/02/2025] [Indexed: 02/10/2025] Open
Abstract
Glioblastoma (GBM), a highly malignant "cold" tumor of the central nervous system, is characterized by its ability to remodel the GBM immune microenvironment (GME), leading to significant resistance to immunotherapy. GBM-associated microglia/macrophages (GAMs) are essential components of the GME. Targeting GAMs has emerged as a promising strategy against GBM. However, their highly immunosuppressive nature contributes to GBM progression and drug resistance, significantly impeding anti-GBM immunotherapy. Accumulating evidence suggests that metabolic reprogramming accompanies GBM progression and GAM polarization, which are in turn driven by specific metabolic abnormalities and altered cellular signaling pathways. Importantly, metabolic crosstalk between GBM and GAMs further promotes tumor progression. Clarifying and disrupting this metabolic crosstalk is expected to enhance the antitumor phenotype of GAMs and inhibit GBM malignant progression. This review explores metabolism-based interregulation between GBM and GAMs and summarizes recent therapeutic strategies targeting this crosstalk, offering new insights into GBM immunotherapy.
Collapse
Affiliation(s)
- Yuan Gao
- Department of Pharmacology, Ningxia Medical University, Yinchuan 750004, China; Hunan Province Key Laboratory for Antibody-Based Drug and Intelligent Delivery System, School of Pharmaceutical Sciences, Hunan University of Medicine, Huaihua 418000, China
| | - Mengxia Zhang
- Department of Histology and Embryology, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Guihua Wang
- Hunan Province Key Laboratory for Antibody-Based Drug and Intelligent Delivery System, School of Pharmaceutical Sciences, Hunan University of Medicine, Huaihua 418000, China
| | - Weiwei Lai
- Hunan Province Key Laboratory for Antibody-Based Drug and Intelligent Delivery System, School of Pharmaceutical Sciences, Hunan University of Medicine, Huaihua 418000, China
| | - Shuxian Liao
- Hunan Province Key Laboratory for Antibody-Based Drug and Intelligent Delivery System, School of Pharmaceutical Sciences, Hunan University of Medicine, Huaihua 418000, China
| | - Yao Chen
- Hunan Province Key Laboratory for Antibody-Based Drug and Intelligent Delivery System, School of Pharmaceutical Sciences, Hunan University of Medicine, Huaihua 418000, China
| | - Qian Ning
- Hunan Province Key Laboratory for Antibody-Based Drug and Intelligent Delivery System, School of Pharmaceutical Sciences, Hunan University of Medicine, Huaihua 418000, China; College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha 410128, China.
| | - Shengsong Tang
- Department of Pharmacology, Ningxia Medical University, Yinchuan 750004, China; Hunan Province Key Laboratory for Antibody-Based Drug and Intelligent Delivery System, School of Pharmaceutical Sciences, Hunan University of Medicine, Huaihua 418000, China; College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha 410128, China.
| |
Collapse
|
13
|
Zhou S, Yang H. Radiotherapy modulates autophagy to reshape the tumor immune microenvironment to enhance anti-tumor immunity in esophageal cancer. Biochim Biophys Acta Rev Cancer 2025; 1880:189302. [PMID: 40120778 DOI: 10.1016/j.bbcan.2025.189302] [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: 07/29/2024] [Revised: 03/15/2025] [Accepted: 03/16/2025] [Indexed: 03/25/2025]
Abstract
The combination of radiotherapy and immunotherapy exerts synergistic antitumor in a range of human cancers, and also in esophageal cancer. Radiotherapy-induced tumor immune microenvironment (TIME) reprogramming is an essential basis for the synergistic antitumor between radiotherapy and immunotherapy. Radiotherapy can induce autophagy in tumor cells and immune cells of TIME, and autophagy activation is involved in the modification of immunological characteristics of TIME. The TIME landscape of esophageal cancer, especially ESCC, can be affected by radiotherapy or autophagy regulation. In this review, we depicted that local radiotherapy-induced autophagy could promote the maturation, migration, infiltration, and function of immune cells by complicated mechanisms to make TIME from immune "cold" to "hot", resulting in the synergistic antitumor of RT and IO. We argue that unraveling the relevance of radiotherapy-initiated autophagy to driving radiotherapy reprogramming TIME will open new ideas to explore new targets or more efficiently multimodal therapeutic interventions in ESCC.
Collapse
Affiliation(s)
- Suna Zhou
- Key Laboratory of Radiation Oncology of Taizhou, Department of Radiation Oncology, Key Laboratory of Minimally Invasive Techniques & Rapid Rehabilitation of Digestive System Tumor of Zhejiang Province, Taizhou Hospital Affiliated to Wenzhou Medical University, Taizhou, Zhejiang 317000, China
| | - Haihua Yang
- Key Laboratory of Radiation Oncology of Taizhou, Department of Radiation Oncology, Key Laboratory of Minimally Invasive Techniques & Rapid Rehabilitation of Digestive System Tumor of Zhejiang Province, Taizhou Hospital Affiliated to Wenzhou Medical University, Taizhou, Zhejiang 317000, China.
| |
Collapse
|
14
|
LOH CHRISTINA, ZHENG YUQI, ALZOUBI ISLAM, ALEXANDER KIMBERLEYL, LEE MAGGIE, CAI WEIDONG, SONG YANG, MCDONALD KERRIE, NOWAK ANNAK, BANATI RICHARDB, GRAEBER MANUELB. Microglia and brain macrophages are differentially associated with tumor necrosis in glioblastoma: A link to tumor progression. Oncol Res 2025; 33:937-950. [PMID: 40191733 PMCID: PMC11964880 DOI: 10.32604/or.2024.056436] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 11/12/2024] [Indexed: 04/09/2025] Open
Abstract
Background Microglia and brain macrophages contribute significantly to the tumor microenvironment in highly malignant glioblastoma where they are considered important drivers of tumor progression. A better understanding of the role of the brain macrophages present in glioblastoma appears crucial for improving therapeutic outcomes, especially in the context of novel immunotherapeutic approaches. Methods We investigated the regulation of two well-established markers for microglia and brain macrophages, IBA1 and CD163, in relation to glioblastoma tumor necrosis using immunohistochemistry and modality fusion heatmaps of whole slide images obtained from adjacent tissue sections. Results IBA1 and CD163 showed remarkable differences in relation to glioblastoma tumor necrosis. Generally, IBA1 immunoreactive cells were far less common in necrotic tissue areas than CD163-expressing cells. We also found extensive and frequently diffuse extracellular CD163 deposition, especially in hypocellular necrobiotic tumor regions where IBA1 was typically absent. Conclusions Resident microglia seem more likely to be important for the diffuse infiltration of glioma cells in hypercellular tissue areas, whereas myeloid macrophages may be the main macrophage population in the wake of tumor necrosis. Since the necrotic niche with its interactions between microglia, brain macrophages, and glioblastoma/glioma stem cells is increasingly recognised as an important factor in tumor progression, further detailed studies of the macrophage populations in glioblastoma are warranted.
Collapse
Affiliation(s)
- CHRISTINA LOH
- Ken Parker Brain Tumor Research Laboratories, Brain and Mind Centre, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2050, Australia
| | - YUQI ZHENG
- Ken Parker Brain Tumor Research Laboratories, Brain and Mind Centre, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2050, Australia
| | - ISLAM ALZOUBI
- School of Computer Science, The University of Sydney, Sydney, NSW 2008, Australia
| | - KIMBERLEY L. ALEXANDER
- Neurosurgery Department, Chris O’Brien Lifehouse, Camperdown, NSW 2050, Australia
- Department of Neuropathology, Royal Prince Alfred Hospital and Brain and Mind Centre, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia
| | - MAGGIE LEE
- Department of Neuropathology, Royal Prince Alfred Hospital and Brain and Mind Centre, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia
| | - WEI-DONG CAI
- School of Computer Science, The University of Sydney, Sydney, NSW 2008, Australia
| | - YANG SONG
- School of Computer Science and Engineering, University of New South Wales, Sydney, NSW 2052, Australia
| | | | - ANNA K. NOWAK
- Medical School, University of Western Australia, Crawley Campus, Perth, WA 6009, Australia
| | - RICHARD B. BANATI
- Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2050, Australia
- Santuario Accademico S. Giovanni D’Andorno, Casa Alpina ‘Principessa Laetitia’, Frazione Bele, Campiglia Cervo, 13812, Italy
| | - MANUEL B. GRAEBER
- Ken Parker Brain Tumor Research Laboratories, Brain and Mind Centre, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2050, Australia
- Department of Neuropathology, Royal Prince Alfred Hospital and Brain and Mind Centre, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia
- University of Sydney Association of Professors (USAP), University of Sydney, Sydney, NSW 2006, Australia
| |
Collapse
|
15
|
Hao Z, Guan W, Wei W, Li M, Xiao Z, Sun Q, Pan Y, Xin W. Unlocking the therapeutic potential of tumor-derived EVs in ischemia-reperfusion: a breakthrough perspective from glioma and stroke. J Neuroinflammation 2025; 22:84. [PMID: 40089793 PMCID: PMC11909855 DOI: 10.1186/s12974-025-03405-7] [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: 01/12/2025] [Accepted: 03/04/2025] [Indexed: 03/17/2025] Open
Abstract
Clinical studies have revealed a bidirectional relationship between glioma and ischemic stroke, with evidence of spatial overlap between the two conditions. This connection arises from significant similarities in their pathological processes, including the regulation of cellular metabolism, inflammation, coagulation, hypoxia, angiogenesis, and neural repair, all of which involve common biological factors. A significant shared feature of both diseases is the crucial role of extracellular vesicles (EVs) in mediating intercellular communication. Extracellular vesicles, with their characteristic bilayer structure, encapsulate proteins, lipids, and nucleic acids, shielding them from enzymatic degradation by ribonucleases, deoxyribonucleases, and proteases. This structural protection facilitates long-distance intercellular communication in multicellular organisms. In gliomas, EVs are pivotal in intracranial signaling and shaping the tumor microenvironment. Importantly, the cargos carried by glioma-derived EVs closely align with the biological factors involved in ischemic stroke, underscoring the substantial impact of glioma on stroke pathology, particularly through the crucial roles of EVs as key mediators in this interaction. This review explores the pathological interplay between glioma and ischemic stroke, addressing clinical manifestations and pathophysiological processes across the stages of hypoxia, stroke onset, progression, and recovery, with a particular focus on the crucial role of EVs and their cargos in these interactions.
Collapse
Affiliation(s)
- Zhongnan Hao
- Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, P. R. China
- Jiangxi Key Laboratory of Neurological Diseases, Department of Neurosurgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
- Department of Neurology, The Affiliated Hospital of Qingdao University, Medical School of Qingdao University, Qingdao, 266100, Shandong Province, China
| | - Wenxin Guan
- Queen Mary School, Nanchang University, Xuefu Avenue, Nanchang, Jiangxi, China
| | - Wei Wei
- Department of Neurology, the Affiliated Hospital of Southwest Jiaotong University & The Third People's Hospital of Chengdu, Chengdu, 610031, Sichuan, PR China
| | - Meihua Li
- Jiangxi Key Laboratory of Neurological Diseases, Department of Neurosurgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Zhipeng Xiao
- Jiangxi Key Laboratory of Neurological Diseases, Department of Neurosurgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Qinjian Sun
- Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, P. R. China
| | - Yongli Pan
- Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, P. R. China.
| | - Wenqiang Xin
- Jiangxi Key Laboratory of Neurological Diseases, Department of Neurosurgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China.
| |
Collapse
|
16
|
Zheng Y, Fuse H, Alzoubi I, Graeber MB. Microglia-Derived Brain Macrophages Associate with Glioblastoma Stem Cells: A Potential Mechanism for Tumor Progression Revealed by AI-Assisted Analysis. Cells 2025; 14:413. [PMID: 40136662 PMCID: PMC11940947 DOI: 10.3390/cells14060413] [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: 01/20/2025] [Revised: 02/21/2025] [Accepted: 03/08/2025] [Indexed: 03/27/2025] Open
Abstract
Background: Malignant gliomas, and notably glioblastoma, are highly aggressive brain tumors. Understanding the mechanisms underlying their progression is crucial for developing more effective treatments. Recent studies have highlighted the role of microglia and brain macrophages in glioblastoma development, but the specific interactions between these immune cells and glioblastoma stem cells (GSCs) remain unclear. Methods: To address this question, we have utilized AI-assisted cell recognition to investigate the spatial relationship between GSCs expressing high levels of CD276 (B7-H3) and microglia- and bone marrow-derived brain macrophages, respectively. Results: Using PathoFusion, our previously developed open-source AI framework, we were able to map specific immunohistochemical phenotypes at the single-cell level within whole-slide images. This approach enabled us to selectively identify Iba1+ and CD163+ macrophages as well as CD276+ GSCs with high specificity and to study their co-localization. Our analysis suggests a closer association of Iba1+ macrophages with GSCs than between CD163+ macrophages and GSCs in glioblastoma. Conclusions: Our findings provide novel insights into the spatial context of tumor immunity in glioblastoma and point to microglia-GSC interactions as a potential mechanism for tumor progression, especially during diffuse tissue infiltration. These findings have significant implications for our understanding of glioblastoma biology, providing a foundation for a comprehensive analysis of microglia activation phenotypes during glioma development. This, in turn, may lead to new therapeutic strategies targeting the early stages of the immune microenvironment of glioblastoma.
Collapse
Affiliation(s)
- Yuqi Zheng
- Ken Parker Brain Tumour Research Laboratories, Brain and Mind Centre, Faculty of Medicine and Health, University of Sydney, Camperdown, Sydney, NSW 2050, Australia;
| | - Haneya Fuse
- School of Medicine, Sydney Campus, University of Notre Dame, 160 Oxford Street, Darlinghurst, Sydney, NSW 2010, Australia;
| | - Islam Alzoubi
- School of Computer Science, The University of Sydney, J12/1 Cleveland St, Darlington, Sydney, NSW 2008, Australia;
| | - Manuel B. Graeber
- Ken Parker Brain Tumour Research Laboratories, Brain and Mind Centre, Faculty of Medicine and Health, University of Sydney, Camperdown, Sydney, NSW 2050, Australia;
- University of Sydney Association of Professors (USAP), University of Sydney, Sydney, NSW 2006, Australia
| |
Collapse
|
17
|
Qian L, Chen P, Zhang S, Wang Z, Guo Y, Koutouratsas V, Fleishman JS, Huang C, Zhang S. The uptake of extracellular vesicles: Research progress in cancer drug resistance and beyond. Drug Resist Updat 2025; 79:101209. [PMID: 39893749 DOI: 10.1016/j.drup.2025.101209] [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: 12/11/2024] [Revised: 01/22/2025] [Accepted: 01/26/2025] [Indexed: 02/04/2025]
Abstract
Extracellular vesicles (EVs) are heterogeneous vesicles released by donor cells that can be taken up by recipient cells, thus inducing cellular phenotype changes. Since their discovery decades ago, roles of EVs in modulating initiation, growth, survival and metastasis of cancer have been revealed. Recent studies from multifaceted perspectives have further detailed the contribution of EVs to cancer drug resistance; however, the role of EV uptake in conferring drug resistance seems to be overlooked. In this comprehensive review, we update the EV subtypes and approaches for determining EV uptake. The biological basis of EV uptake is systematically summarized. Moreover, we focus on the diverse uptake mechanisms by which EVs carry out the intracellular delivery of functional molecules and drug resistance signaling. Furthermore, we highlight how EV uptake confers drug resistance and identify potential strategies for targeting EV uptake to overcome drug resistance. Finally, we discuss the research gap on the role of EV uptake in promoting drug resistance. This updated knowledge provides a new avenue to overcome cancer drug resistance by targeting EV uptake.
Collapse
Affiliation(s)
- Luomeng Qian
- Department of Cell Biology, School of Medicine, Nankai University, Tianjin, 300071, China
| | - Pangzhou Chen
- Department of Breast Surgery, Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Foshan 528200, China
| | - Shiwu Zhang
- Department of Pathology, Tianjin Union Medical Center, Nankai University, Tianjin 300121, China
| | - Zhenglu Wang
- Department of Pathology, Tianjin Key Laboratory for Organ Transplantation, Tianjin First Centre Hospital, Tianjin 300192, China
| | - Yuan Guo
- Department of Cell Biology, School of Medicine, Nankai University, Tianjin, 300071, China
| | - Vasili Koutouratsas
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, USA
| | - Joshua S Fleishman
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, USA
| | - Chuanqiang Huang
- Department of Breast Surgery, Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Foshan 528200, China
| | - Sihe Zhang
- Department of Cell Biology, School of Medicine, Nankai University, Tianjin, 300071, China.
| |
Collapse
|
18
|
Li X, Liu X, Gao Y, Li L, Wang Y, Men J, Ren J, Wang J, Li F, Li Y, Xiong J, Cui X, Wei C, Wang C, Dong J, Liu L, Zhang J, Zhang S. Glioblastoma Cells Express and Secrete Alternatively Spliced Transcripts of Coagulation Factor X. Biomedicines 2025; 13:576. [PMID: 40149552 PMCID: PMC11940189 DOI: 10.3390/biomedicines13030576] [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: 01/09/2025] [Revised: 02/19/2025] [Accepted: 02/20/2025] [Indexed: 03/29/2025] Open
Abstract
Background: Patients with cancer often develop a prothrombotic state that can evolve into venous and arterial thrombosis, which is associated with poor clinical outcomes. Glioblastoma multiforme (GBM) is the most frequently associated with thrombosis, but the underlying causes of this prothrombotic state are poorly defined. Objectives: We designed a study to characterize the expression of coagulation factor X (FX) and its alternatively spliced transcripts in glioblastoma tissues surgically removed from patients and in clonal cell lines. Methods: The F10 mRNA and FX protein were quantified in tissues surgically removed from seven patients with glioblastoma (glioma grade 3-4) and those from non-tumor patients. Glioblastoma cells from three human clonal lines were examined for the expression and secretion of FX at baseline and after the cells were stimulated with lipopolysaccharide (LPS) or subjected to oxygen/glucose starvation in culture. PCR products were subjected to Sanger sequencing and amplicon sequencing to identify F10 isoforms and their ratios. A chromogenic assay was performed to assess FX activity. Results: Glioblastoma tissue and cell lines expressed high levels of the full-length and an alternatively spliced F10 mRNA. The latter produced a C-terminal truncated FX. The ratio of full-length to truncated F10 transcripts was significantly higher in normal brain tissues than in glioblastoma tissue. In cultured cells from the glioblastoma cell lines, FX was secreted to the conditioned medium and was active in cleaving a chemical substrate. The FX expression and secretion were upregulated in cells stimulated with LPS or subjected to oxygen/glucose starvation. Discussion: Glioblastoma cells synthesize and secrete FX that was active in promoting thrombin generation. These findings provide a new underlying mechanism to explain why glioblastoma patients are prone to developing thrombosis.
Collapse
Affiliation(s)
- Xiaotian Li
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300052, China; (X.L.); (Y.G.); (L.L.); (F.L.); (Y.L.); (J.X.); (X.C.); (C.W.); (C.W.)
- Key Laboratory of Post-Neuro-Injury Repair and Regeneration in Central Nervous System, Tianjin Neurological Institute, Tianjin 300052, China;
| | - Xilei Liu
- Department of Urology, Tianjin Medical University General Hospital, Tianjin 300052, China;
| | - Yalong Gao
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300052, China; (X.L.); (Y.G.); (L.L.); (F.L.); (Y.L.); (J.X.); (X.C.); (C.W.); (C.W.)
- Key Laboratory of Post-Neuro-Injury Repair and Regeneration in Central Nervous System, Tianjin Neurological Institute, Tianjin 300052, China;
| | - Lei Li
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300052, China; (X.L.); (Y.G.); (L.L.); (F.L.); (Y.L.); (J.X.); (X.C.); (C.W.); (C.W.)
- Key Laboratory of Post-Neuro-Injury Repair and Regeneration in Central Nervous System, Tianjin Neurological Institute, Tianjin 300052, China;
| | - Yajuan Wang
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin 300052, China;
| | - Jianlong Men
- Center for Precision Medicine, Tianjin Medical University General Hospital, Tianjin 300052, China; (J.M.); (J.R.)
| | - Jing Ren
- Center for Precision Medicine, Tianjin Medical University General Hospital, Tianjin 300052, China; (J.M.); (J.R.)
| | - Jiwei Wang
- Department of Neurosurgery, Tianjin Huanhu Hospital, Tianjin 300350, China;
| | - Fanjian Li
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300052, China; (X.L.); (Y.G.); (L.L.); (F.L.); (Y.L.); (J.X.); (X.C.); (C.W.); (C.W.)
- Key Laboratory of Post-Neuro-Injury Repair and Regeneration in Central Nervous System, Tianjin Neurological Institute, Tianjin 300052, China;
| | - Yaohua Li
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300052, China; (X.L.); (Y.G.); (L.L.); (F.L.); (Y.L.); (J.X.); (X.C.); (C.W.); (C.W.)
| | - Jianhua Xiong
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300052, China; (X.L.); (Y.G.); (L.L.); (F.L.); (Y.L.); (J.X.); (X.C.); (C.W.); (C.W.)
| | - Xiaoteng Cui
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300052, China; (X.L.); (Y.G.); (L.L.); (F.L.); (Y.L.); (J.X.); (X.C.); (C.W.); (C.W.)
- Key Laboratory of Post-Neuro-Injury Repair and Regeneration in Central Nervous System, Tianjin Neurological Institute, Tianjin 300052, China;
| | - Cheng Wei
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300052, China; (X.L.); (Y.G.); (L.L.); (F.L.); (Y.L.); (J.X.); (X.C.); (C.W.); (C.W.)
- Key Laboratory of Post-Neuro-Injury Repair and Regeneration in Central Nervous System, Tianjin Neurological Institute, Tianjin 300052, China;
| | - Cong Wang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300052, China; (X.L.); (Y.G.); (L.L.); (F.L.); (Y.L.); (J.X.); (X.C.); (C.W.); (C.W.)
- Key Laboratory of Post-Neuro-Injury Repair and Regeneration in Central Nervous System, Tianjin Neurological Institute, Tianjin 300052, China;
| | - Jingfei Dong
- Bloodworks Research Institute, Division of Hematology, Department of Medicine, University of Washington School of Medicine, Seattle, WA 98195, USA;
| | - Li Liu
- Key Laboratory of Post-Neuro-Injury Repair and Regeneration in Central Nervous System, Tianjin Neurological Institute, Tianjin 300052, China;
| | - Jianning Zhang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300052, China; (X.L.); (Y.G.); (L.L.); (F.L.); (Y.L.); (J.X.); (X.C.); (C.W.); (C.W.)
- Key Laboratory of Post-Neuro-Injury Repair and Regeneration in Central Nervous System, Tianjin Neurological Institute, Tianjin 300052, China;
| | - Shu Zhang
- Key Laboratory of Post-Neuro-Injury Repair and Regeneration in Central Nervous System, Tianjin Neurological Institute, Tianjin 300052, China;
| |
Collapse
|
19
|
Wang Q, Shi Y, Qin Z, Xu M, Wang J, Lu Y, Zhao Z, Bi H. A375 melanoma-derived lactate controls A375 melanoma phenotypes by inducing macrophage M2 polarization via TCA cycle and TGF-β signaling. PeerJ 2025; 13:e18887. [PMID: 39995996 PMCID: PMC11849511 DOI: 10.7717/peerj.18887] [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: 01/31/2024] [Accepted: 12/31/2024] [Indexed: 02/26/2025] Open
Abstract
Introduction Macrophage phenotypes have been linked to progression and prognosis of cutaneous melanoma. However, the association between Warburg effect in A375 melanoma and macrophages polarization, as well as the underlying mechanisms, remains less well documented. Objective The present study aimed to investigate the effect of lactate derived from A375 melanoma on macrophage polarization, melanoma phenotype responses and the underlying mechanisms. Methods Flow cytometry was performed to evaluate the expression of M1 and M2 markers, cell cycle and apoptosis. Levels of transforming growth factor β (TGF-β) and tumor necrosis factor α (TNF-α) were determined with enzyme-linked immunosorbent assay (ELISA) kit. Proliferation and invasion were assessed by CCK8 and transwell assays, respectively. The extracellular acidification rate (ECAR) and oxygen consumption rate (OCR) were analyzed using an XF96 extracellular flux analyzer. Protein expressions were determined by Western blotting. Results Our results revealed that melanoma A375 conditioned medium (A375-CM) induced peripheral blood mononuclear cells (PBMCs) to polarize toward anti-inflammatory M2 macrophages. M2 markers CD206 and ARG1 expression increased, as did TGF-β secretion. Conversely, M1 marker CD68 expression decreased. Furthermore, hypoxia promoted macrophage M2 polarization induced by A375-CM. Elevated lactate level in PIG1-conditioned medium (PIG1-CM) induced M2 polarization, whereas the lactate transport inhibitor AZD3965 suppressed this effect in PBMCs cultured with A375-CM. Additionally, lactate derived from melanoma regulated M1/M2 polarization by the tricarboxylic acid (TCA) cycle instead of glycolysis. Significantly, polarized macrophages altered melanoma phenotypes including proliferation, clone formation, cell cycle, apoptosis, migration and invasion via TCA cycle and TGF-β. Conclusion Our data collectively demonstrate that lactate derived from melanoma facilitates polarization of M2 macrophages, which subsequently leads to modifications in melanoma phenotypes via TCA cycle and TGF-β signaling.
Collapse
Affiliation(s)
- Qifei Wang
- Department of Plastic Surgery, Peking University Third Hospital, Haidian District, Beijing, China
| | - Yurui Shi
- Department of Plastic Surgery, Peking University Third Hospital, Haidian District, Beijing, China
| | - Zelian Qin
- Department of Plastic Surgery, Peking University Third Hospital, Haidian District, Beijing, China
| | - Mengli Xu
- Department of Plastic Surgery, Peking University Third Hospital, Haidian District, Beijing, China
| | - Jingyi Wang
- Department of Plastic Surgery, Peking University Third Hospital, Haidian District, Beijing, China
| | - Yuhao Lu
- Department of Plastic Surgery, Peking University Third Hospital, Haidian District, Beijing, China
| | - Zhenmin Zhao
- Department of Plastic Surgery, Peking University Third Hospital, Haidian District, Beijing, China
| | - Hongsen Bi
- Department of Plastic Surgery, Peking University Third Hospital, Haidian District, Beijing, China
| |
Collapse
|
20
|
Yang H, Xue Y, Jiang Q, Tian Q, Xu J, Li J, Yang Q, Du M, Yang T, Wei X, Zhao M, Yan T, Chen X, Li L. HSPA5-mediated glioma hypoxia tolerance promotes M2 macrophage polarization under hypoxic microenvironment. Int Immunopharmacol 2025; 147:113856. [PMID: 39740502 DOI: 10.1016/j.intimp.2024.113856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 12/10/2024] [Accepted: 12/11/2024] [Indexed: 01/02/2025]
Abstract
BACKGROUND The tumor microenvironment (TME), with hallmark features of hypoxia and immunosuppression, plays a crucial role in the progression of various solid tumors. However, the intricate interplay between tumor hypoxia and the formation of tumor immune microenvironment in glioma remains incompletely understood. METHODS In the present study, we initially identified genes associated with tumor hypoxia and the immune microenvironment through GSEA and IMMPORT database analysis. We subsequently identified hypoxia- and immune-related genes associated with glioma prognosis through further cross-analysis and multidatabase integrated analysis. HSPA5 was ultimately identified as a potential target gene related to the formation of the hypoxic microenvironment and immune microenvironment in glioma. Furthermore, we conducted MTT, colony formation, EdU, migration and invasion assays and intracranial orthotopic tumor model analysis to further evaluate the impact of interfering with HSPA5 expression on the hypoxic and immune microenvironments of glioma. RESULTS We found that HSPA5 is highly expressed in glioma cells and tissues and is associated with a poor prognosis. Further investigation revealed that hypoxia promotes the malignant biological characteristics of glioma and reshaping the Immunosuppressive phenotype of tumor-associated macrophages (TAMs) through upregulation of the HIF-1α/HSPA5 axis. Silencing HSPA5 alleviated glioma hypoxia tolerance and induced the polarization of TAMs toward the M1 phenotype. The induced macrophages could exhibit a tumor-suppressive effect. CONCLUSION These observations suggest that HSPA5 upregulation promotes glioma progression by inducing hypoxia tolerance and reshaping the Immunosuppressive phenotype of TAMs. Therefore, targeting HSPA5 may be a novel therapeutic strategy for glioma.
Collapse
Affiliation(s)
- He Yang
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang Province, China; Key Colleges and Universities Laboratory of Neurosurgery in Heilongjiang Province, Harbin 150001, Heilongjiang Province, China; Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, Harbin 150001, Heilongjiang Province, China
| | - Yanpeng Xue
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang Province, China; Key Colleges and Universities Laboratory of Neurosurgery in Heilongjiang Province, Harbin 150001, Heilongjiang Province, China; Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, Harbin 150001, Heilongjiang Province, China
| | - Qing Jiang
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang Province, China; Key Colleges and Universities Laboratory of Neurosurgery in Heilongjiang Province, Harbin 150001, Heilongjiang Province, China; Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, Harbin 150001, Heilongjiang Province, China
| | - Qingqing Tian
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang Province, China; Key Colleges and Universities Laboratory of Neurosurgery in Heilongjiang Province, Harbin 150001, Heilongjiang Province, China; Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, Harbin 150001, Heilongjiang Province, China
| | - Jiayi Xu
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang Province, China; Key Colleges and Universities Laboratory of Neurosurgery in Heilongjiang Province, Harbin 150001, Heilongjiang Province, China; Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, Harbin 150001, Heilongjiang Province, China
| | - Jixuan Li
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang Province, China; Key Colleges and Universities Laboratory of Neurosurgery in Heilongjiang Province, Harbin 150001, Heilongjiang Province, China; Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, Harbin 150001, Heilongjiang Province, China
| | - Quan Yang
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang Province, China; Key Colleges and Universities Laboratory of Neurosurgery in Heilongjiang Province, Harbin 150001, Heilongjiang Province, China; Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, Harbin 150001, Heilongjiang Province, China
| | - Mingdong Du
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang Province, China; Key Colleges and Universities Laboratory of Neurosurgery in Heilongjiang Province, Harbin 150001, Heilongjiang Province, China; Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, Harbin 150001, Heilongjiang Province, China
| | - Teng Yang
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang Province, China; Key Colleges and Universities Laboratory of Neurosurgery in Heilongjiang Province, Harbin 150001, Heilongjiang Province, China; Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, Harbin 150001, Heilongjiang Province, China
| | - Xingwang Wei
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang Province, China; Key Colleges and Universities Laboratory of Neurosurgery in Heilongjiang Province, Harbin 150001, Heilongjiang Province, China; Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, Harbin 150001, Heilongjiang Province, China
| | - Mei Zhao
- Department of Pharmacy, Sanya Central Hospital (The Third People's Hospital of Hainan Province), Sanya 572000, China
| | - Tao Yan
- Central Laboratory, Linyi People's Hospital, Linyi 276000, Shandong Province, China; Linyi Key Laboratory of Neurophysiology, Linyi People's Hospital, Linyi 276000, Shandong Province, China.
| | - Xin Chen
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang Province, China; Key Colleges and Universities Laboratory of Neurosurgery in Heilongjiang Province, Harbin 150001, Heilongjiang Province, China; Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, Harbin 150001, Heilongjiang Province, China.
| | - Lixian Li
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang Province, China; Key Colleges and Universities Laboratory of Neurosurgery in Heilongjiang Province, Harbin 150001, Heilongjiang Province, China; Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, Harbin 150001, Heilongjiang Province, China.
| |
Collapse
|
21
|
Li J, Zhao Y, Wu X, Zou Y, Liu Y, Ma H. Choline kinase alpha regulates autophagy-associated exosome release to promote glioma cell progression. Biochem Biophys Res Commun 2025; 746:151269. [PMID: 39778250 DOI: 10.1016/j.bbrc.2024.151269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 12/30/2024] [Accepted: 12/30/2024] [Indexed: 01/11/2025]
Abstract
Glioma is the most common primary intracranial malignant tumor in adults, with a poor prognosis. Exosomes released by tumor cells play a crucial role in tumor development, metastasis, angiogenesis, and other biological processes. Despite this significance, the precise molecular mechanisms governing exosome secretion and their impact on tumor progression remain incompletely understood. While Choline Kinase Alpha (CHKA) has been implicated in promoting various types of tumors, its specific role in glioma pathogenesis remains unclear. Our study initially demonstrates that CHKA enhances the proliferation, migration, and invasion abilities of glioma cells. Interestingly, CHKA also stimulates the release of exosomes from glioma cells. Mechanistically, reduced CHKA expression hampers exosome secretion by elevating autophagy levels in gliomas, whereas counteracting the autophagy elevation resulting from CHKA downregulation restores the release of exosomes. Notably, exosomes derived from glioma cells with normal CHKA expression exhibit a greater capacity to promote glioma progression compared to those derived from cells with low CHKA expression. Overall, our findings suggest that CHKA modulates exosome secretion via an autophagy-dependent pathway, thereby facilitating the proliferation, migration, and invasion of glioma cells.
Collapse
Affiliation(s)
- Jialin Li
- School of Clinical Medicine, Ningxia Medical University, Yinchuan, China
| | - Yang Zhao
- School of Clinical Medicine, Ningxia Medical University, Yinchuan, China
| | - Xiao Wu
- School of Clinical Medicine, Ningxia Medical University, Yinchuan, China
| | - Yourui Zou
- Department of Neurosurgery, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Yang Liu
- Department of Neurosurgery, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Hui Ma
- Department of Neurosurgery, General Hospital of Ningxia Medical University, Yinchuan, China.
| |
Collapse
|
22
|
Peppicelli S, Calorini L, Bianchini F, Papucci L, Magnelli L, Andreucci E. Acidity and hypoxia of tumor microenvironment, a positive interplay in extracellular vesicle release by tumor cells. Cell Oncol (Dordr) 2025; 48:27-41. [PMID: 39023664 PMCID: PMC11850579 DOI: 10.1007/s13402-024-00969-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/28/2024] [Indexed: 07/20/2024] Open
Abstract
The complex and continuously evolving features of the tumor microenvironment, varying between tumor histotypes, are characterized by the presence of host cells and tumor cells embedded in a milieu shaped by hypoxia and low pH, resulting from the frequent imbalance between vascularity and tumor cell proliferation. These microenvironmental metabolic stressors play a crucial role in remodeling host cells and tumor cells, contributing to the stimulation of cancer cell heterogeneity, clonal evolution, and multidrug resistance, ultimately leading to progression and metastasis. The extracellular vesicles (EVs), membrane-enclosed structures released into the extracellular milieu by tumor/host cells, are now recognized as critical drivers in the complex intercellular communication between tumor cells and the local cellular components in a hypoxic/acidic microenvironment. Understanding the intricate molecular mechanisms governing the interactions between tumor and host cells within a hypoxic and acidic microenvironment, triggered by the release of EVs, could pave the way for innovative strategies to disrupt the complex interplay of cancer cells with their microenvironment. This approach may contribute to the development of an efficient and safe therapeutic strategy to combat cancer progression. Therefore, we review the major findings on the release of EVs in a hypoxic/acidic tumor microenvironment to appreciate their role in tumor progression toward metastatic disease.
Collapse
Affiliation(s)
- Silvia Peppicelli
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, 50134, Italy.
| | - Lido Calorini
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, 50134, Italy
| | - Francesca Bianchini
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, 50134, Italy
| | - Laura Papucci
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, 50134, Italy
| | - Lucia Magnelli
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, 50134, Italy
| | - Elena Andreucci
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, 50134, Italy
| |
Collapse
|
23
|
Zhang J, Hu S, Jin X, Zheng Y, Yu L, Ma J, Gu B, Wang F, Wu W. Hypoxia-Associated GPNMB+ Macrophages Promote Malignant Progression of Colorectal Cancer and Its Related Risk Signature Are Powerful Predictive Tool for the Treatment of Colorectal Cancer Patients. ENVIRONMENTAL TOXICOLOGY 2025; 40:204-221. [PMID: 39367576 DOI: 10.1002/tox.24426] [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: 02/07/2024] [Revised: 07/03/2024] [Accepted: 07/23/2024] [Indexed: 10/06/2024]
Abstract
Colorectal cancer (CRC) is a highly malignant tumor with hypoxia being a crucial feature during its progression. This study utilized multiple independent CRC cohorts for bioinformatics analysis and in vitro experiments to investigate the role of hypoxia-related subgroups in CRC. Machine learning was employed to construct risk features associated with this subgroup and further explore its therapeutic value in CRC. The study identified the GPNMB+ Macrophage (GPNMB+ Macr) subgroup as most relevant to hypoxia. GPNMB+ Macr showed significantly higher infiltration in tumor tissues compared to non-tumor tissues, increasing with CRC stage. High infiltration of GPNMB+ Macr was associated with poor prognosis in terms of overall and recurrence-free survival in CRC patients. GPNMB+ Macrophages exhibit M2-like characteristics and have the ability to promote 5-FU resistance, proliferation, and metastasis of CRC cells. The study developed the Hypoxia-Related Macrophage Risk Score (HMRS), which not only served as an independent prognostic factor for CRC patients but also demonstrated robust prognostic performance compared to 84 previously published prognostic features. Patients with low HMRS were sensitive to fluorouracil, oxaliplatin (FOLFOX), and anti-PD-1 immunotherapy, while those with high HMRS showed resistance. Additionally, HMRS was identified as an independent prognostic factor in other digestive tract tumors (hepatocellular carcinoma, pancreatic cancer, esophageal cancer, and gastric cancer), indicating potential extrapolation to other tumor types. In conclusion, GPNMB+ Macr promotes the malignant progression of CRC, and HMRS serves as a powerful predictive tool for prognosis, chemotherapy, and immunotherapy in CRC patients, aiding in improving the quality of survival.
Collapse
Affiliation(s)
- Junli Zhang
- Department of Blood Transfusion, The Third People's Hospital of Bengbu Affiliated to Bengbu Medical University, Bengbu, Anhui, China
- Department of Biochemistry and Molecular Biology, Bengbu Medical University Key Laboratory of Cancer Research and Clinical Laboratory Diagnosis, Bengbu Medical University, Bengbu, Anhui, China
| | - Shangshang Hu
- Department of Biochemistry and Molecular Biology, Bengbu Medical University Key Laboratory of Cancer Research and Clinical Laboratory Diagnosis, Bengbu Medical University, Bengbu, Anhui, China
- Department of Clinical Medicine, School of Medicine, Southeast University, Nanjing, Jiangsu, China
| | - Xinxin Jin
- Department of Biochemistry and Molecular Biology, Bengbu Medical University Key Laboratory of Cancer Research and Clinical Laboratory Diagnosis, Bengbu Medical University, Bengbu, Anhui, China
| | - Yiwen Zheng
- Department of Biochemistry and Molecular Biology, Bengbu Medical University Key Laboratory of Cancer Research and Clinical Laboratory Diagnosis, Bengbu Medical University, Bengbu, Anhui, China
| | - Lianchen Yu
- Department of Biochemistry and Molecular Biology, Bengbu Medical University Key Laboratory of Cancer Research and Clinical Laboratory Diagnosis, Bengbu Medical University, Bengbu, Anhui, China
| | - Junrao Ma
- Department of Biochemistry and Molecular Biology, Bengbu Medical University Key Laboratory of Cancer Research and Clinical Laboratory Diagnosis, Bengbu Medical University, Bengbu, Anhui, China
| | - Biao Gu
- Department of Blood Transfusion, The Third People's Hospital of Bengbu Affiliated to Bengbu Medical University, Bengbu, Anhui, China
| | - Fen Wang
- Department of Blood Transfusion, The Third People's Hospital of Bengbu Affiliated to Bengbu Medical University, Bengbu, Anhui, China
| | - Wenjuan Wu
- Department of Biochemistry and Molecular Biology, School of Laboratory Medicine, Bengbu Medical College, Bengbu, China
| |
Collapse
|
24
|
Branco H, Xavier CPR, Riganti C, Vasconcelos MH. Hypoxia as a critical player in extracellular vesicles-mediated intercellular communication between tumor cells and their surrounding microenvironment. Biochim Biophys Acta Rev Cancer 2025; 1880:189244. [PMID: 39672279 DOI: 10.1016/j.bbcan.2024.189244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 12/06/2024] [Accepted: 12/09/2024] [Indexed: 12/15/2024]
Abstract
In the past years, increasing attention has been paid to the role of extracellular vesicles (EVs) as mediators of intercellular communication in cancer. These small size particles mediate the intercellular transfer of important bioactive molecules involved in malignant initiation and progression. Hypoxia, or low partial pressure of oxygen, is recognized as a remarkable feature of solid tumors and has been demonstrated to exert a profound impact on tumor prognosis and therapeutic efficacy. Indeed, the high-pitched growth rate and chaotic neovascular architecture that embodies solid tumors results in a profound reduction in oxygen pressure within the tumor microenvironment (TME). In response to oxygen-deprived conditions, tumor cells and their surrounding milieu develop homeostatic adaptation mechanisms that contribute to the establishment of a pro-tumoral phenotype. Latest evidence suggests that the hypoxic microenvironment that surrounds the tumor bulk may be a clincher for the observed elevated levels of circulating EVs in cancer patients. Thus, it is proposed that EVs may play a role in mediating intercellular communication in response to hypoxic conditions. This review focuses on the EVs-mediated crosstalk that is established between tumor cells and their surrounding immune, endothelial, and stromal cell populations, within the hypoxic TME.
Collapse
Affiliation(s)
- Helena Branco
- i3S - Instituto de Investigação e Inovação em Saúde, University of Porto, 4200-135 Porto, Portugal; Cancer Drug Resistance Group, IPATIMUP - Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135 Porto, Portugal; Department of Biological Sciences, FFUP - Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Cristina P R Xavier
- i3S - Instituto de Investigação e Inovação em Saúde, University of Porto, 4200-135 Porto, Portugal; Cancer Drug Resistance Group, IPATIMUP - Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135 Porto, Portugal; UCIBIO - Applied Molecular Biosciences Unit, Toxicologic Pathology Research Laboratory, University Institute of Health Sciences (1H-TOXRUN, IUCS-CESPU), 4585-116 Gandra, Portugal; Associate Laboratory i4HB - Institute for Health and Bioeconomy, University Institute of Health Sciences - CESPU, 4585-116 Gandra, Portugal.
| | - Chiara Riganti
- Department of Oncology, University of Torino, 10126 Torino, Italy; Interdepartmental Research Center for Molecular Biotechnology "G. Tarone", University of Torino, 10126 Torino, Italy
| | - M Helena Vasconcelos
- i3S - Instituto de Investigação e Inovação em Saúde, University of Porto, 4200-135 Porto, Portugal; Cancer Drug Resistance Group, IPATIMUP - Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135 Porto, Portugal; Department of Biological Sciences, FFUP - Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal.
| |
Collapse
|
25
|
Dalpati N, Rai SK, Sharma P, Sarangi PP. Integrins and integrin-driven secretory pathways as multi-dimensional regulators of tumor-associated macrophage recruitment and reprogramming in tumor microenvironment. Matrix Biol 2025; 135:55-69. [PMID: 39645091 DOI: 10.1016/j.matbio.2024.12.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 11/27/2024] [Accepted: 12/04/2024] [Indexed: 12/09/2024]
Abstract
Integrins, a group of transmembrane receptors, play a crucial role in mediating the interactions between cells and extracellular matrix (ECM) proteins. The intracellular signaling initiated by these cell-matrix interactions in leukocytes mediates many essential cellular processes such as survival, migration, metabolism, and other immunological functions. Macrophages, as phagocytes, participate in both proinflammatory and anti-inflammatory processes, including progression. Numerous reports have shown that the integrin-regulated secretome, comprising cytokines, chemokines, growth factors, proteases, and other bioactive molecules, is a crucial modulator of macrophage functions in tumors, significantly influencing macrophage programming and reprogramming within the tumor microenvironment (TME) in addition to driving their step-by-step entry process into tumor tissue spaces. Importantly, studies have demonstrated a pivotal role for integrin receptor-mediated secretome and associated signaling pathways in functional reprogramming from anti-tumorigenic to pro-tumorigenic phenotype in tumor-associated macrophages (TAMs). In this comprehensive review, we have provided an in-depth analysis of the latest findings of various key pathways, mediators, and signaling cascades associated with integrin-driven polarization of macrophages in tumors. This manuscript will provide an updated understanding of the modulation of inflammatory monocytes/ macrophages and TAMs by integrin-driven secretory pathways in various functions such as migration, differentiation, and their role in tumor progression, angiogenesis, and metastasis.
Collapse
Affiliation(s)
- Nibedita Dalpati
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee 247667, Uttarakhand, India
| | - Shubham Kumar Rai
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee 247667, Uttarakhand, India
| | - Prerna Sharma
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee 247667, Uttarakhand, India
| | - Pranita P Sarangi
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee 247667, Uttarakhand, India.
| |
Collapse
|
26
|
Li X, Zhao H. Targeting secretory autophagy in solid cancers: mechanisms, immune regulation and clinical insights. Exp Hematol Oncol 2025; 14:12. [PMID: 39893499 PMCID: PMC11786567 DOI: 10.1186/s40164-025-00603-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Accepted: 01/25/2025] [Indexed: 02/04/2025] Open
Abstract
Secretory autophagy is a classical form of unconventional secretion that integrates autophagy with the secretory process, relying on highly conserved autophagy-related molecules and playing a critical role in tumor progression and treatment resistance. Traditional autophagy is responsible for degrading intracellular substances by fusing autophagosomes with lysosomes. However, secretory autophagy uses autophagy signaling to mediate the secretion of specific substances and regulate the tumor microenvironment (TME). Cytoplasmic substances are preferentially secreted rather than directed toward lysosomal degradation, involving various selective mechanisms. Moreover, substances released by secretory autophagy convey biological signals to the TME, inducing immune dysregulation and contributing to drug resistance. Therefore, elucidating the mechanisms underlying secretory autophagy is essential for improving clinical treatments. This review systematically summarizes current knowledge of secretory autophagy, from initiation to secretion, considering inter-tumor heterogeneity, explores its role across different tumor types. Furthermore, it proposes future research directions and highlights unresolved clinical challenges.
Collapse
Affiliation(s)
- Xinyu Li
- Department of General Surgery, Fourth Affiliated Hospital of China Medical University, Shenyang City, 110032, Liaoning Province, China
| | - Haiying Zhao
- Department of General Surgery, Fourth Affiliated Hospital of China Medical University, Shenyang City, 110032, Liaoning Province, China.
| |
Collapse
|
27
|
Liu L, An Z, Zhang H, Wan X, Zhao X, Yang X, Tian J, Song X. Bone marrow mesenchymal stem cell-derived extracellular vesicles alleviate diabetes-exacerbated atherosclerosis via AMPK/mTOR pathway-mediated autophagy-related macrophage polarization. Cardiovasc Diabetol 2025; 24:48. [PMID: 39881287 PMCID: PMC11780875 DOI: 10.1186/s12933-025-02603-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Accepted: 01/17/2025] [Indexed: 01/31/2025] Open
Abstract
INTRODUCTION Bone marrow-derived mesenchymal stem cell-derived extracellular vesicles (BMSC-EVs) are widely used for therapeutic purposes in preclinical studies. However, their utility in treating diabetes-associated atherosclerosis remains largely unexplored. Here, we aimed to characterize BMSC-EV-mediated regulation of autophagy and macrophage polarization. METHODS EVs were isolated from the supernatant of cultured BMSCs and characterized with transmission electron microscopy (TEM), nanoparticle tracking analysis (NTA), and western blotting. A diabetes-related atherosclerotic ApoE-/- mouse model was established through feeding with a high-fat diet (HFD) and streptozotocin (STZ). Histopathological analyses were carried out using Oil Red O, H&E, and Masson staining of the aorta. TEM and immunohistochemistry (IHC) were applied to evaluate autophagy, and immunofluorescence (IF) was used to identify macrophage polarization. RAW264.7 macrophages were induced with oxidized low-density lipoprotein (ox-LDL) and high glucose (HG), co-cultured with BMSC-EVs, and analyzed for macrophage proliferation, migration, and foam cell formation. RAW264.7 cells were transduced with autophagy marker mRFP-GFP-LC3 lentivirus and analyzed with IF and western blotting. RESULTS Diabetic mice (DA group) had larger aortic plaque areas and lower collagen content than the HFD mice. BMSC-EV treatment significantly reduced blood glucose, LDL levels, and aortic plaque areas while increasing collagen content. BMSC-EV-treated aortas contained a higher number of autophagosomes/autolysosomes, with increased expression of LC3BII correlating with decreased P62 levels and a lower proportion of M1 macrophages. In vitro, BMSC-EVs inhibited proliferation, migration, and foam cell formation in ox-LDL and HG-induced activated RAW264.7 cells. These effects were reversed by the autophagy blocker bafilomycin A1. Consistent with the in vivo findings, BMSC-EVs elevated levels of the autophagy-related protein LC3BII/I and decreased P62 in ox-LDL and HG-induced RAW264.7 cells. These cells also expressed the M1 macrophage markers CD86 and iNOS, but showed reduced expression of the M2 marker Arg-1. Further, BMSC-EVs decreased AMPKα and mTOR phosphorylation levels, which were blocked by the AMPK inhibitor compound C. CONCLUSIONS BMSC-EVs attenuate diabetes-exacerbated atherosclerosis by inhibiting vascular macrophage proliferation, migration, and foam cell formation via AMPK/mTOR signaling-regulated autophagy and macrophage polarization. BMSC-EVs thus hold promise as therapeutic agents for atherosclerosis.
Collapse
Affiliation(s)
- Libo Liu
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, No.2 Anzhen Road, Chaoyang District, 100029, Beijing, China
- Department of Cardiology, The Second Affiliated Hospital of Shandong First Medical University, No. 366 Taishan Street, Taishan District, 271000, Tai'an, China
| | - Ziyu An
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, No.2 Anzhen Road, Chaoyang District, 100029, Beijing, China
| | - Huan Zhang
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, No.2 Anzhen Road, Chaoyang District, 100029, Beijing, China
| | - Xueqi Wan
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, No.2 Anzhen Road, Chaoyang District, 100029, Beijing, China
| | - Xin Zhao
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, No.2 Anzhen Road, Chaoyang District, 100029, Beijing, China
| | - Xueyao Yang
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, No.2 Anzhen Road, Chaoyang District, 100029, Beijing, China
| | - Jinfan Tian
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, No.2 Anzhen Road, Chaoyang District, 100029, Beijing, China.
| | - Xiantao Song
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, No.2 Anzhen Road, Chaoyang District, 100029, Beijing, China.
| |
Collapse
|
28
|
Liu X, Zhao J, Sun Q, Xue Z, Tang Z, Liu W, Liu J, Miao B, Su N, He Y, Zhu Y, Huang B, Yang N, Li C, Wang J, Wang X. Calnexin promotes glioblastoma progression by inducing protective mitophagy through the MEK/ERK/BNIP3 pathway. Theranostics 2025; 15:2624-2648. [PMID: 39990231 PMCID: PMC11840740 DOI: 10.7150/thno.105591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 01/08/2025] [Indexed: 02/25/2025] Open
Abstract
Rationale: Glioblastoma multiforme (GBM), one of the most malignant tumors of the central nervous system, has a poor prognosis, mainly because of its high recurrence caused by the rapid development of drug resistance to postoperative chemotherapy. Although macroautophagy/autophagy is believed to be a fundamental factor in tumor survival during chemotherapy, there is still a lack of autophagy biomarkers for predicting patient prognosis and chemotherapeutic efficacy in clinical practice. Methods: We combined transcriptomic and single-cell sequencing data to identify differentially expressed autophagy-related genes in gliomas. Overexpression of calnexin (CANX), a key gene related to protein folding, and its secretion in the endoplasmic reticulum (ER) was identified, suggesting poor prognosis in GBM patients. The autophagy flow related to CANX was detected by transmission electron microscopy (TEM), Western blotting, and immunofluorescence. Flow cytometry, cell proliferation, activity assays, and the GBM intracranial xenograft mouse model were employed to validate CANX's role in GBM progression. Results: CANX knockdown inhibited proliferation and autophagosome formation in GBM cells. On the other hand, CANX overexpression increased mitogen-activated protein kinase (MAPK) activity, leading to the accumulation of BNIP3 (CL2/adenovirus E1B 19 kDa interacting protein 3, a critical factor regulating mitophagy) and protective mitophagy. Notably, when combined with temozolomide (TMZ), CANX knockdown extended the lifespan of GBM-bearing mice. Additionally, our studies revealed that the classic calcium inhibitor nimodipine (ND) decreased CANX expression and thus enhanced the sensitivity to TMZ. Conclusions: Our findings indicate that CANX functions as an oncogene in GBM. We also characterize the CANX/MEK/ERK/BNIP3 mitophagy pathway, provide new insights into the molecular mechanism of GBM drug resistance, and identify a therapeutic target.
Collapse
Affiliation(s)
- Xuchen Liu
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, 250012 Jinan, China
- Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Function Remodeling, 250117 Jinan, China
- School of Medicine, Cheeloo College of Medicine, Shandong University, 250012 Jinan, China
| | - Jiangli Zhao
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, 250012 Jinan, China
- Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Function Remodeling, 250117 Jinan, China
- School of Medicine, Cheeloo College of Medicine, Shandong University, 250012 Jinan, China
| | - Qingyuan Sun
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, 250012 Jinan, China
- Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Function Remodeling, 250117 Jinan, China
| | - Zhiwei Xue
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, 250012 Jinan, China
- Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Function Remodeling, 250117 Jinan, China
- School of Medicine, Cheeloo College of Medicine, Shandong University, 250012 Jinan, China
| | - Ziyi Tang
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, 250012 Jinan, China
- Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Function Remodeling, 250117 Jinan, China
| | - Wenyu Liu
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, 250012 Jinan, China
- Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Function Remodeling, 250117 Jinan, China
- School of Medicine, Cheeloo College of Medicine, Shandong University, 250012 Jinan, China
| | - Junzhi Liu
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, 250012 Jinan, China
- Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Function Remodeling, 250117 Jinan, China
| | - Baojian Miao
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, 250012 Jinan, China
- Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Function Remodeling, 250117 Jinan, China
- School of Medicine, Cheeloo College of Medicine, Shandong University, 250012 Jinan, China
| | - Nan Su
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, 250012 Jinan, China
- Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Function Remodeling, 250117 Jinan, China
- School of Medicine, Cheeloo College of Medicine, Shandong University, 250012 Jinan, China
| | - Yanya He
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, 250012 Jinan, China
- Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Function Remodeling, 250117 Jinan, China
- School of Medicine, Cheeloo College of Medicine, Shandong University, 250012 Jinan, China
| | - Yuehua Zhu
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, 250012 Jinan, China
- Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Function Remodeling, 250117 Jinan, China
- School of Medicine, Cheeloo College of Medicine, Shandong University, 250012 Jinan, China
| | - Bin Huang
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, 250012 Jinan, China
- Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Function Remodeling, 250117 Jinan, China
- School of Medicine, Cheeloo College of Medicine, Shandong University, 250012 Jinan, China
| | - Ning Yang
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, 250012 Jinan, China
- Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Function Remodeling, 250117 Jinan, China
- School of Medicine, Cheeloo College of Medicine, Shandong University, 250012 Jinan, China
| | - Chao Li
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, 250012 Jinan, China
- Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Function Remodeling, 250117 Jinan, China
- School of Medicine, Cheeloo College of Medicine, Shandong University, 250012 Jinan, China
| | - Jiwei Wang
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, 250012 Jinan, China
- Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Function Remodeling, 250117 Jinan, China
- School of Medicine, Cheeloo College of Medicine, Shandong University, 250012 Jinan, China
| | - Xinyu Wang
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, 250012 Jinan, China
- Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Function Remodeling, 250117 Jinan, China
- School of Medicine, Cheeloo College of Medicine, Shandong University, 250012 Jinan, China
| |
Collapse
|
29
|
Shen Y, Lin J, Jiang T, Shen X, Li Y, Fu Y, Xu P, Fang L, Chen Z, Huang H, Xia Y, Xu Z, Wang L. GC-derived exosomal circMAN1A2 promotes cancer progression and suppresses T-cell antitumour immunity by inhibiting FBXW11-mediated SFPQ degradation. J Exp Clin Cancer Res 2025; 44:24. [PMID: 39856764 PMCID: PMC11762487 DOI: 10.1186/s13046-025-03288-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Accepted: 01/13/2025] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND Exosomes, as extracellular membrane vesicles, play important roles in intercellular communication and can influence tumour progression. Circular RNAs (circRNAs) have been reported in various malignancies and are also important components of exosomes. However, the role of exosomal circRNAs in gastric cancer (GC) progression has not been completely clarified. METHODS The exosomal circRNAs enriched in GC were identified using exosomal circRNA sequencing. The biological function of circMAN1A2 in GC was investigated using a series of in vitro and in vivo experiments. PKH-67 staining was used to label the exosomes. The molecular mechanism of exosomal circMAN1A2 was investigated via mass spectrometry, immunoprecipitation, Western blot, and single-cell RNA-sequencing data analyses. RESULTS In our study, we determined that circMAN1A2 (hsa_circ_0000118) was enriched in GC-derived exosomes. Higher circMAN1A2 expression was related to poor survival in GC patients (HR = 2.917, p = 0.0120). Exosomal circMAN1A2 promoted GC progression in vitro and in vivo and suppressed the antitumour activity of T cells. Moreover, circMAN1A2 bound to SFPQ in GC cells and T cells, promoting the G1/S phase transition of the cell cycle in GC cells while inhibiting the activation of the T cell receptor signalling pathway in T cells to decrease antitumour activity. Mechanistically, circMAN1A2 competed with FBXW11 for binding to SFPQ, preventing FBXW11-mediated k48-linked ubiquitination and SFPQ protein degradation, thereby stabilizing SFPQ expression. CONCLUSIONS Our work confirms the critical role of exosomal circMAN1A2 in the progression and immunosuppression of GC. This novel axis of circMAN1A2-SFPQ provides new insights into exosomal circRNA-based GC diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- Yikai Shen
- Gastric Cancer Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Jie Lin
- Gastric Cancer Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Tianlu Jiang
- The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi Medical Center, Nanjing Medical University, Wuxi People's Hospital, Wuxi, Jiangsu Province, China
| | - Xusheng Shen
- Gastric Cancer Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Ying Li
- Gastric Cancer Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Yiwang Fu
- Gastric Cancer Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Penghui Xu
- Gastric Cancer Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Lang Fang
- Gastric Cancer Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Zetian Chen
- Gastric Cancer Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Hongxin Huang
- Gastric Cancer Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Yiwen Xia
- Gastric Cancer Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China.
| | - Zekuan Xu
- Gastric Cancer Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China.
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu Province, China.
| | - Linjun Wang
- Gastric Cancer Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China.
| |
Collapse
|
30
|
Zhang J, Zhang J, Yang C. Autophagy in brain tumors: molecular mechanisms, challenges, and therapeutic opportunities. J Transl Med 2025; 23:52. [PMID: 39806481 PMCID: PMC11727735 DOI: 10.1186/s12967-024-06063-0] [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: 07/20/2024] [Accepted: 12/27/2024] [Indexed: 01/16/2025] Open
Abstract
Autophagy is responsible for maintaining cellular balance and ensuring survival. Autophagy plays a crucial role in the development of diseases, particularly human cancers, with actions that can either promote survival or induce cell death. However, brain tumors contribute to high levels of both mortality and morbidity globally, with resistance to treatments being acquired due to genetic mutations and dysregulation of molecular mechanisms, among other factors. Hence, having knowledge of the role of molecular processes in the advancement of brain tumors is enlightening, and the current review specifically examines the role of autophagy. The discussion would focus on the molecular pathways that control autophagy in brain tumors, and its dual role as a tumor suppressor and a supporter of tumor survival. Autophagy can control the advancement of different types of brain tumors like glioblastoma, glioma, and ependymoma, demonstrating its potential for treatment. Autophagy mechanisms can influence metastasis and drug resistance in glioblastoma, and there is a complex interplay between autophagy and cellular responses to stress like hypoxia and starvation. Autophagy can inhibit the growth of brain tumors by promoting apoptosis. Hence, focusing on autophagy could offer fresh perspectives on creating successful treatments.
Collapse
Affiliation(s)
- Jiarui Zhang
- Department of Pathology, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Jinan Zhang
- Department of Neurosurgery, Tangdu Hospital, Fourth Military Medical University, No. 569 Xinsi Road, Xi'an, China.
| | - Chen Yang
- Department of Neurosurgery, Tangdu Hospital, Fourth Military Medical University, No. 569 Xinsi Road, Xi'an, China.
| |
Collapse
|
31
|
Liu Y, Tan H, Dai J, Lin J, Zhao K, Hu H, Zhong C. Targeting macrophages in cancer immunotherapy: Frontiers and challenges. J Adv Res 2025:S2090-1232(24)00622-2. [PMID: 39778768 DOI: 10.1016/j.jare.2024.12.043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 12/28/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Cancer immunotherapy has emerged as a groundbreaking approach in cancer treatment, primarily realized through the manipulation of immune cells, notably T cell adoption and immune checkpoint blockade. Nevertheless, the manipulation of T cells encounters formidable hurdles. Macrophages, serving as the pivotal link between innate and adaptive immunity, play crucial roles in phagocytosis, cytokine secretion, and antigen presentation. Consequently, macrophage-targeted therapies have garnered significant attention. AIM OF REVIEW We aim to provide the most cutting-edge insights and future perspectives for macrophage-targeted therapies, fostering the development of novel and effective cancer treatments. KEY SCIENTIFIC CONCEPTS OF REVIEW To date, the forefront strategies for macrophage targeting encompass: altering their plasticity, harnessing CAR-macrophages, and targeting phagocytosis checkpoints. Macrophages are characterized by their remarkable diversity and plasticity, offering a unique therapeutic target. In this context, we critically analyze the innovative strategies aimed at transforming macrophages from their M2 (tumor-promoting) to M1 (tumor-suppressing) phenotype. Furthermore, we delve into the design principles, developmental progress, and advantages of CAR-macrophages. Additionally, we illuminate the challenges encountered in targeting phagocytosis checkpoints on macrophages and propose potential strategies to overcome these obstacles.
Collapse
Affiliation(s)
- Yu'e Liu
- Department of Neurosurgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200120, China; Department of Pediatric Hematology-Oncology, Boston Children's Hospital, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA.
| | - Huabing Tan
- Department of Infectious Diseases, Hepatology Institute, Renmin Hospital, Hubei University of Medicine, Shiyan Key Laboratory of Virology, Hubei University of Medicine, Shiyan, Hubei Province 442000, China; General internal medicine, Wuhan Jinyintan Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, 430048, China
| | - Jingyuan Dai
- School of Computer Science and Information Systems, Northwest Missouri State University, Maryville, MO 64468, USA
| | - Jianghua Lin
- Department of Neurosurgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200120, China
| | - Kaijun Zhao
- Department of Neurosurgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200120, China.
| | - Haibo Hu
- Department of Cardiothoracic Surgery, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an, Jiangsu, China.
| | - Chunlong Zhong
- Department of Neurosurgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200120, China.
| |
Collapse
|
32
|
Tripathi S, Sharma Y, Kumar D. Biological Cargo: Exosomes and their Role in Cancer Progression and Metastasis. Curr Top Med Chem 2025; 25:263-285. [PMID: 38984577 DOI: 10.2174/0115680266304636240626055711] [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: 01/16/2024] [Revised: 05/23/2024] [Accepted: 05/23/2024] [Indexed: 07/11/2024]
Abstract
Cancer cells are among the many types of cells that release exosomes, which are nanovesicles. Because of their many potential applications, exosomes have recently garnered much attention from cancer researchers. The bioactive substances that exosomes release as cargo have been the subject of several investigations. The substances in question may operate as biomarkers for diagnosis or affect apoptosis, the immune system, the development and spread of cancer, and other processes. Others have begun to look at exosomes in experimental therapeutic trials because they believe they may be useful in the treatment of cancer. This review started with a short description of exosome biogenesis and key features. Next, the potential of tumor-derived exosomes and oncosomes to influence the immune system throughout the development of cancer, as well as alter tumor microenvironments (TMEs) and pre-metastatic niche creation, was investigated. Finally, there was talk of exosomes' possible use in cancer treatment. Furthermore, there is emerging consensus about the potential application of exosomes to be biological reprogrammers of cancer cells, either as carriers of naturally occurring chemicals, including anticancer medications, or as carriers of anticancer vaccines for immunotherapy as well as boron neutron capture therapy (BNCT). We briefly review the key ideas and logic behind this intriguing therapy recommendation.
Collapse
Affiliation(s)
- Siddhant Tripathi
- Poona College of Pharmacy, Bharati Vidyapeeth (Deemed to be) University, Pune, Maharashtra, 411038, India
| | - Yashika Sharma
- Poona College of Pharmacy, Bharati Vidyapeeth (Deemed to be) University, Pune, Maharashtra, 411038, India
| | - Dileep Kumar
- Poona College of Pharmacy, Bharati Vidyapeeth (Deemed to be) University, Pune, Maharashtra, 411038, India
| |
Collapse
|
33
|
Laurenge A, Castro-Vega LJ, Huberfeld G. Reciprocal interactions between glioma and tissue-resident cells fueling tumor progression. HANDBOOK OF CLINICAL NEUROLOGY 2025; 210:177-190. [PMID: 40148044 DOI: 10.1016/b978-0-443-19102-2.00007-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/29/2025]
Abstract
Gliomas are the most frequent primary brain tumor and are essentially incurable. While nondiffuse gliomas are circumscribed, diffuse gliomas display an aggressive behavior characterized by tumor cell migration over large distances into the brain parenchyma, thereby precluding curative surgical resection. Almost all diffuse gliomas progress and recur as higher grades and become resistant to standard-of-care treatments. It is being increasingly recognized that glioma cells establish functional interactions with cells residing in the tumor microenvironment. Of these, tumor-associated microglia and macrophages (TAMs) play critical roles in immunosuppression through modulation of the extracellular matrix, and the secretion of molecules such as cytokines, neurotrophic factors, and micro-RNAs (miRNAs). Conversely, glioma cell signals influence cell states and drive the metabolic reprogramming of TAMs. Similarly, emergent evidence indicates that neuronal activity influences glioma by released factors and by establishing functional synapses with glioma cells to promote tumor growth and invasion. Glioma cells also affect local neuronal activities and maintain connections through microtube gap junctions to amplify local effects. Here, we discuss the molecular mechanisms underlying bidirectional interactions between glioma cells and TAMs, as well as between glioma cells and neurons. A better understanding of these cellular cross talks is crucial for the development of novel therapeutic strategies for diffuse gliomas.
Collapse
Affiliation(s)
- Alice Laurenge
- Genetics & Development of Brain Tumors Laboratory, ICM - Paris Brain Institute, Sorbonne University, UMR S 1127, Inserm U 1127, CNRS UMR 7225, F-75013, Paris, France; AP-HP, Groupe Hospitalier Pitié-Salpêtrière, Neuro-Oncology Department, F-75013, Paris, France
| | - Luis Jaime Castro-Vega
- Genetics & Development of Brain Tumors Laboratory, ICM - Paris Brain Institute, Sorbonne University, UMR S 1127, Inserm U 1127, CNRS UMR 7225, F-75013, Paris, France
| | - Gilles Huberfeld
- Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, Neuronal Signaling in Epilepsy and Glioma, Paris, France; Department of Neurology, Hôpital Fondation Adolphe de Rothschild, Paris, France.
| |
Collapse
|
34
|
Wang H, Jia L, Yu H, Tang H, Chi H, Zhang W, Chen J. Mechanism Study of Bufalin Reversal of Drug Resistance by Inhibiting Hypoxic Colon Cancer Cell-Induced Polarization of M2 Macrophages. Integr Cancer Ther 2025; 24:15347354251325806. [PMID: 40071641 PMCID: PMC11898227 DOI: 10.1177/15347354251325806] [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: 08/10/2024] [Revised: 01/24/2025] [Accepted: 02/20/2025] [Indexed: 03/15/2025] Open
Abstract
Chemoresistance is still an important factor affecting the efficacy of treatment in colorectal cancer (CRC) patients. Hypoxia is related to poor prognosis and treatment resistance in cancer. Relevant studies have shown that a hypoxic microenvironment can promote the polarization of M2 macrophages and thus promote tumor development. Previous research has found that bufalin has a wide range of antitumor effects, but whether bufalin can reverse tumor resistance by improving the hypoxic tumor microenvironment is still unclear. In present research, it was found that high expression of SRC-3 in CRC cells under hypoxic conditions promoted the polarization of M2 and caused chemotherapy resistance, while bufalin, a monomeric drug used in Chinese medicine, reduced the level of SRC-3 and HIF-1α, thereby reversing chemoresistance. In addition, overexpression of SRC-3 reduced the hypoxia-mitigating effect of bufalin on CRC cells to promote the polarization of M2. Bufalin also inhibits the polarization of M2 caused by hypoxic CRC cells. Therefore, bufalin has the potential to become a new adjuvant therapy that can be further explored in future studies on its treatment of CRC.
Collapse
Affiliation(s)
- Haijing Wang
- Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Linlin Jia
- Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hongjie Yu
- Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hui Tang
- Chongqing Medical and Pharmaceutical College, Chongqing, China
| | - Huabowen Chi
- Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wei Zhang
- Three Gorges University & Yichang Hospital of Traditional Chinese Medicine, Yichang, China
| | - Jinbao Chen
- Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
35
|
Gao L, Jing X, Hua Q, Li Z, Lei P, Song P, Zhou L, Tian Y, Liu J, Cai Q. Complement C1S is a potential prognostic biomarker and associated with M2 macrophage infiltration in gliomas: From bioinformatics to comprehensive experimental validation. Int Immunopharmacol 2024; 143:113573. [PMID: 39515040 DOI: 10.1016/j.intimp.2024.113573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Revised: 11/02/2024] [Accepted: 11/03/2024] [Indexed: 11/16/2024]
Abstract
Glioma is the most common malignant tumor of the central nervous system, and the ability of traditional clinical treatment to prolong the survival of glioma patients is limited. A substantial body of evidence underscores the pivotal role of the immune system in eradicating malignant cells and impeding tumor metastasis. Consequently, tumor immunotherapy has become a promising avenue to address the clinical conundrum faced by glioma patients. The complement system is a natural immune system that is an important line of defense in the immune response. C1S plays a key role in activating the classical complement system. Nevertheless, few studies have focused on the role of C1S in glioma tumorigenesis and progression. In this study, we demonstrated that C1S was upregulated in GBM (Grade IV) and low-grade gliomas (LGG, Grade II-III) by combining glioma cohorts from multiple public databases with our internal independent cohorts and that increased C1S expression levels predict a poor prognosis for gliomas. Cox regression analysis identified C1S as an important prognostic indicator for glioma patients. In addition, gene functional enrichment analysis demonstrated that C1S was involved in cellular immunity, T-cell activation, macrophage differentiation, and cell proliferation. Further experiments demonstrated that C1S facilitates tumor cell proliferation, cell migration and intracranial tumor growth in nude mice. More importantly, we evaluated the role of C1S in immune infiltration. These results suggested that C1S was closely related to a variety of immune cell types in glioma, especially M2 macrophages. Our findings were further validated via glioma tissue microarray immunohistochemical analysis and an M2 macrophage infiltration assay. Together, these findings revealed the underlying critical role of C1S in glioma tumorigenesis, progression, and the tumor immune microenvironment, contributing to further understanding of glioma pathogenesis and guiding immunotherapy.
Collapse
Affiliation(s)
- Lun Gao
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, PR China; Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, PR China
| | - Xiongfei Jing
- Department of Neurosurgery, Xiantao First People's Hospital Affiliated to Yangtze University, Xiantao City 433000, PR China
| | - Qiuwei Hua
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, PR China; Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, PR China
| | - Zhiyang Li
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, PR China; Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, PR China
| | - Pan Lei
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, PR China; Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, PR China
| | - Ping Song
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, PR China; Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, PR China
| | - Long Zhou
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, PR China; Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, PR China
| | - Yihao Tian
- Department of Human Anatomy, School of Basic Medical Sciences, Wuhan University, Wuhan, Hubei 430071, PR China.
| | - Junhui Liu
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, PR China; Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, PR China.
| | - Qiang Cai
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, PR China; Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, PR China.
| |
Collapse
|
36
|
Bajaj N, Sharma D. Uncovering metabolic signatures in cancer-derived exosomes: LC-MS/MS and NMR profiling. NANOSCALE 2024; 17:287-303. [PMID: 39565062 DOI: 10.1039/d4nr03454f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2024]
Abstract
Understanding the intricate interplay between cancer metabolism and intercellular communication within the tumour microenvironment (TME) is crucial for advancing cancer diagnostics and therapeutics. In this study, we investigate the metabolites present in exosomes derived from three distinct cancer cell lines: pancreatic cancer (MiaPaCa-2), lung cancer (A549), and glioma (C6). Exosomes were isolated using ultrafiltration and characterized using a combination of techniques including nanoparticle tracking analysis (NTA), electron microscopy (EM), western blotting (WB) and Fourier-transform infrared (FTIR) spectroscopy. Leveraging state-of-the-art metabolomics techniques, including untargeted LC-MS/MS and NMR analyses, we elucidated the metabolic signatures encapsulated within cancer-derived exosomes. Notably, our investigation represents the first exploration of exosomal metabolites from pancreatic and glioma cells, addressing a significant gap in current knowledge. Furthermore, our study investigates the correlation between metabolites derived from different cancer cells, shedding light on potential metabolic interactions within the TME. Through comprehensive analyses, this study provides insights into dysregulated metabolic pathways driving cancer progression and offers novel perspectives on the diagnostic and therapeutic utility of exosomal metabolites. Importantly, common metabolites identified among cancer types suggest potential markers detectable by multiple techniques, enhancing their clinical applicability.
Collapse
Affiliation(s)
- Nandini Bajaj
- Institute of Nano Science and Technology, Knowledge City, Sector 81, Mohali, Punjab, 140306, India.
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Deepika Sharma
- Institute of Nano Science and Technology, Knowledge City, Sector 81, Mohali, Punjab, 140306, India.
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| |
Collapse
|
37
|
Pilotto Heming C, Aran V. The potential of circulating cell-free RNA in CNS tumor diagnosis and monitoring: A liquid biopsy approach. Crit Rev Oncol Hematol 2024; 204:104504. [PMID: 39251048 DOI: 10.1016/j.critrevonc.2024.104504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 08/20/2024] [Accepted: 09/04/2024] [Indexed: 09/11/2024] Open
Abstract
Early detection of malignancies, through regular cancer screening, has already proven to have potential to increase survival rates. Yet current screening methods rely on invasive, expensive tissue sampling that has hampered widespread use. Liquid biopsy is noninvasive and represents a potential approach to precision oncology, based on molecular profiling of body fluids. Among these, circulating cell-free RNA (cfRNA) has gained attention due to its diverse composition and potential as a sensitive biomarker. This review provides an overview of the processes of cfRNA delivery into the bloodstream and the role of cfRNA detection in the diagnosis of central nervous system (CNS) tumors. Different types of cfRNAs such as microRNAs (miRNAs), long noncoding RNAs (lncRNAs) and circular RNAs (circRNAs) have been recognized as potential biomarkers in CNS tumors. These molecules exhibit differential expression patterns in the plasma, cerebrospinalfluid (CSF) and urine of patients with CNS tumors, providing information for diagnosing the disease, predicting outcomes, and assessing treatment effectiveness. Few clinical trials are currently exploring the use of liquid biopsy for detecting and monitoring CNS tumors. Despite obstacles like sample standardization and data analysis, cfRNA shows promise as a tool in the diagnosis and management of CNS tumors, offering opportunities for early detection, personalized therapy, and improved patient outcomes.
Collapse
Affiliation(s)
- Carlos Pilotto Heming
- Programa de Pós-Graduação em Anatomia Patológica, Faculdade de Medicina, Universidade Federal do Rio de Janeiro (UFRJ), Av. Rodolpho Paulo Rocco 225, Rio de Janeiro 21941-905, Brazil; Instituto Estadual do Cérebro Paulo Niemeyer (IECPN), Rua do Rezende 156, Rio de Janeiro 20231-092, Brazil
| | - Veronica Aran
- Programa de Pós-Graduação em Anatomia Patológica, Faculdade de Medicina, Universidade Federal do Rio de Janeiro (UFRJ), Av. Rodolpho Paulo Rocco 225, Rio de Janeiro 21941-905, Brazil; Instituto Estadual do Cérebro Paulo Niemeyer (IECPN), Rua do Rezende 156, Rio de Janeiro 20231-092, Brazil.
| |
Collapse
|
38
|
Huang L, Zhan J, Li Y, Huang K, Zhu X, Li J. The roles of extracellular vesicles in gliomas: Challenge or opportunity? Life Sci 2024; 358:123150. [PMID: 39471898 DOI: 10.1016/j.lfs.2024.123150] [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: 01/26/2024] [Revised: 07/07/2024] [Accepted: 10/14/2024] [Indexed: 11/01/2024]
Abstract
Gliomas are increasingly becoming a major disease affecting human health, and current treatments are not as effective as expected. Deeper insights into glioma heterogeneity and the search for new diagnostic and therapeutic strategies appear to be urgent. Gliomas adapt to their surroundings and form a supportive tumor microenvironment (TME). Glioma cells will communicate with the surrounding cells through extracellular vesicles (EVs) carrying bioactive substances such as nucleic acids, proteins and lipids which is related to the modification to various metabolic pathways and regulation of biological behaviors, and this regulation can be bidirectional, widely existing between cells in the TME, constituting a complex network of interactions. This complex regulation can affect glioma therapy, leading to different types of resistance. Because of the feasibility of EVs isolation in various body fluids, they have a promising usage in the diagnosis and monitoring of gliomas. At the same time, the nature of EVs to cross the blood-brain barrier (BBB) confers potential for their use as drug delivery systems. In this review, we will focus on the roles and functions of EVs derived from different cellular origins in the glioma microenvironment and the intercellular regulatory networks, and explore possible clinical applications in glioma diagnosis and precision therapy.
Collapse
Affiliation(s)
- Le Huang
- Department of Neurosurgery, The 2nd Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, PR China; HuanKui Academy, Jiangxi Medical College, Nanchang University, Nanchang 330031, China
| | - Jianhao Zhan
- HuanKui Academy, Jiangxi Medical College, Nanchang University, Nanchang 330031, China
| | - Yao Li
- The 1st affiiated hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi, PR China
| | - Kai Huang
- Department of Neurosurgery, The 2nd Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, PR China; Institute of Neuroscience, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, PR China; Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang, China; JXHC Key Laboratory of Neurological Medicine, Jiangxi 330006, Nanchang, PR China.
| | - Xingen Zhu
- Department of Neurosurgery, The 2nd Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, PR China; Institute of Neuroscience, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, PR China; Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang, China; JXHC Key Laboratory of Neurological Medicine, Jiangxi 330006, Nanchang, PR China
| | - Jingying Li
- Department of Comprehensive Intensive Care Unit, The 2nd Affiliated Hospital, Jiangxi Medical University, Nanchang University, Nanchang, PR China.
| |
Collapse
|
39
|
Wu J, Lei JH, Li M, Zhang A, Li Y, Liang X, de Souza SC, Yuan Z, Wang C, Chen G, Liu TM, Deng CX, Tang Z, Qu S. Carbon Dots Crosslinked Egg White Hydrogel for Tissue Engineering. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2404702. [PMID: 39303206 DOI: 10.1002/advs.202404702] [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: 05/01/2024] [Revised: 09/01/2024] [Indexed: 09/22/2024]
Abstract
Egg white (EW)-derived hydrogels hold promise as biomaterials for in vitro cell culture due to their ability to mimic the extracellular matrix. However, their highly cross-linked structures restrict their potential for in vivo applications, as they are unable to integrate dynamically with tissues before degradation. In this study, this limitation is addressed by introducing carbon dots (CDs) as cross-linking agents for EW in a dilute aqueous solution. The resulting CDs-crosslinked EW hydrogel (CEWH) exhibits tensile strength comparable to that of skin tissue and features a large pore structure that promotes cell infiltration. Subcutaneous implantation of CEWH demonstrated excellent integration with surrounding tissue and a degradation rate aligned with the hair follicles (HFs) regeneration cycle. This allows the long-term regeneration and establishment of an M2 macrophage-dominated immune microenvironment, which in turn promotes the re-entry of HFs into the anagen phase from the telogen phase. Additionally, CEWH demonstrated potential as a wound dressing material. Overall, this study paves the way for utilizing EW as a versatile biomaterial for tissue engineering.
Collapse
Affiliation(s)
- Jun Wu
- Joint Key Laboratory of the Ministry of Education, Institute of Applied Physics and Materials Engineering, University of Macau, Macau, 999078, P. R. China
- Department of Physics and Chemistry, Faculty of Science and Technology University of Macau, Macao, 999078, P. R. China
- MOE Frontier Science Centre for Precision Oncology University of Macau, Macao, 999078, P. R. China
| | - Josh Haipeng Lei
- MOE Frontier Science Centre for Precision Oncology University of Macau, Macao, 999078, P. R. China
- Faculty of Health Sciences, University of Macau, Macau, 999078, P. R. China
| | - Moxin Li
- MOE Frontier Science Centre for Precision Oncology University of Macau, Macao, 999078, P. R. China
- Faculty of Health Sciences, University of Macau, Macau, 999078, P. R. China
| | - Aiping Zhang
- MOE Frontier Science Centre for Precision Oncology University of Macau, Macao, 999078, P. R. China
- Faculty of Health Sciences, University of Macau, Macau, 999078, P. R. China
| | - Yuan Li
- Joint Key Laboratory of the Ministry of Education, Institute of Applied Physics and Materials Engineering, University of Macau, Macau, 999078, P. R. China
- Department of Physics and Chemistry, Faculty of Science and Technology University of Macau, Macao, 999078, P. R. China
- MOE Frontier Science Centre for Precision Oncology University of Macau, Macao, 999078, P. R. China
| | - Xiao Liang
- MOE Frontier Science Centre for Precision Oncology University of Macau, Macao, 999078, P. R. China
- Faculty of Health Sciences, University of Macau, Macau, 999078, P. R. China
| | - Senio Campos de Souza
- Institute of Chinese Medical Sciences & State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macau, 999078, P. R. China
| | - Zhen Yuan
- MOE Frontier Science Centre for Precision Oncology University of Macau, Macao, 999078, P. R. China
- Faculty of Health Sciences, University of Macau, Macau, 999078, P. R. China
| | - Chunming Wang
- Institute of Chinese Medical Sciences & State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macau, 999078, P. R. China
| | - Guokai Chen
- MOE Frontier Science Centre for Precision Oncology University of Macau, Macao, 999078, P. R. China
- Faculty of Health Sciences, University of Macau, Macau, 999078, P. R. China
| | - Tzu-Ming Liu
- MOE Frontier Science Centre for Precision Oncology University of Macau, Macao, 999078, P. R. China
- Faculty of Health Sciences, University of Macau, Macau, 999078, P. R. China
| | - Chu-Xia Deng
- MOE Frontier Science Centre for Precision Oncology University of Macau, Macao, 999078, P. R. China
- Faculty of Health Sciences, University of Macau, Macau, 999078, P. R. China
| | - Zikang Tang
- Joint Key Laboratory of the Ministry of Education, Institute of Applied Physics and Materials Engineering, University of Macau, Macau, 999078, P. R. China
- Department of Physics and Chemistry, Faculty of Science and Technology University of Macau, Macao, 999078, P. R. China
- MOE Frontier Science Centre for Precision Oncology University of Macau, Macao, 999078, P. R. China
| | - Songnan Qu
- Joint Key Laboratory of the Ministry of Education, Institute of Applied Physics and Materials Engineering, University of Macau, Macau, 999078, P. R. China
- Department of Physics and Chemistry, Faculty of Science and Technology University of Macau, Macao, 999078, P. R. China
- MOE Frontier Science Centre for Precision Oncology University of Macau, Macao, 999078, P. R. China
| |
Collapse
|
40
|
Peng T, Chai M, Chen Z, Wu M, Li X, Han F, Chen S, Liao C, Yue M, Song YQ, Wu H, Tian L, An G. Exosomes from Hypoxia Preconditioned Muscle-Derived Stem Cells Enhance Cell-Free Corpus Cavernosa Angiogenesis and Reproductive Function Recovery. Adv Healthc Mater 2024; 13:e2401406. [PMID: 39007245 DOI: 10.1002/adhm.202401406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 06/24/2024] [Indexed: 07/16/2024]
Abstract
Tissue engineering for penile corpora cavernosa defects requires microvascular system reconstruction.GelMA hydrogels show promise for tissue regeneration. However, using stem cells faces challenges such as immune rejection, limited proliferation and differentiation, and biosafety concerns. Therefore, acellular tissue regeneration may avoid these issues. Exosomes are used from muscle-derived stem cells (MDSCs) to modify 3D-printed hydrogel scaffolds for acellular tissue regeneration. Hypoxia-preconditioned MDSC-derived exosomes are obtained to enhance the therapeutic effect. In contrast to normoxic exosomes (N-Exos), hypoxic exosomes (H-Exos) are found to markedly enhance the proliferation, migration, and capillary-like tube formation of human umbilical vein endothelial cells (HUVECs). High-throughput sequencing analysis of miRNAs isolated from both N-Exos and H-Exos revealed a significant upregulation of miR-21-5p in H-Exos following hypoxic preconditioning. Further validation demonstrated that the miR-21-5p/PDCD4 pathway promoted the proliferation of HUVECs. Epigallocatechin gallate (EGCG) is introduced to improve the mechanical properties and biocompatibility of GelMA hydrogels. EGCG-GelMA scaffolds loaded with different types of Exos are transplanted to repair rabbit penile corpora cavernosa defects, observed the blood flow and repair status of the defect site through color Doppler ultrasound and magnetic resonance imaging, and ultimately restored the rabbit penile erection function and successfully bred offspring. Thus, acellular hydrogel scaffolds offer an effective treatment for penile corpora cavernosa defects.
Collapse
Affiliation(s)
- Tianwen Peng
- Department of Obstetrics and Gynecology, Center of Reproductive Medicine, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, Guangdong-Hong Kong-Macao Greater Bay Area Higher Education Joint Laboratory Maternal-Fetal Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, P. R. China
| | - Muyuan Chai
- National Engineering Research Centre for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, P. R. China
| | - Zhicong Chen
- Department of Obstetrics and Gynecology, Center of Reproductive Medicine, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, Guangdong-Hong Kong-Macao Greater Bay Area Higher Education Joint Laboratory Maternal-Fetal Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, P. R. China
| | - Man Wu
- Department of Obstetrics and Gynecology, Center of Reproductive Medicine, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, Guangdong-Hong Kong-Macao Greater Bay Area Higher Education Joint Laboratory Maternal-Fetal Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, P. R. China
| | - Xiaomin Li
- Department of Obstetrics and Gynecology, Center of Reproductive Medicine, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, Guangdong-Hong Kong-Macao Greater Bay Area Higher Education Joint Laboratory Maternal-Fetal Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, P. R. China
| | - Feixue Han
- Department of Obstetrics and Gynecology, Center of Reproductive Medicine, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, Guangdong-Hong Kong-Macao Greater Bay Area Higher Education Joint Laboratory Maternal-Fetal Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, P. R. China
| | - Shuyan Chen
- Department of Obstetrics and Gynecology, Center of Reproductive Medicine, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, Guangdong-Hong Kong-Macao Greater Bay Area Higher Education Joint Laboratory Maternal-Fetal Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, P. R. China
| | - Chen Liao
- Department of Obstetrics and Gynecology, Center of Reproductive Medicine, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, Guangdong-Hong Kong-Macao Greater Bay Area Higher Education Joint Laboratory Maternal-Fetal Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, P. R. China
| | - Ming Yue
- School of Biomedical Sciences, AIDS Institute and Department of Microbiology, The University of Hong Kong, Hong Kong, 999077, P. R. China
| | - You-Qiang Song
- School of Biomedical Sciences, The State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Hong Kong, 999077, P. R. China
| | - Hongkai Wu
- Department of Chemistry, The Hong Kong University of Science and Technology, Hong Kong, 999077, P. R. China
| | - Long Tian
- Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, P. R. China
| | - Geng An
- Department of Obstetrics and Gynecology, Center of Reproductive Medicine, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, Guangdong-Hong Kong-Macao Greater Bay Area Higher Education Joint Laboratory Maternal-Fetal Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, P. R. China
| |
Collapse
|
41
|
Yuan H, Li Y, Kong Z, Peng L, Song J, Hou X, Zhang W, Liu R, Feng T, Zhu C. IL-33-Pretreated Mesenchymal Stem Cells Attenuate Acute Liver Failure by Improving Homing and Polarizing M2 Macrophages. Stem Cells Int 2024; 2024:1273099. [PMID: 39478979 PMCID: PMC11524710 DOI: 10.1155/2024/1273099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Revised: 07/05/2024] [Accepted: 09/18/2024] [Indexed: 11/02/2024] Open
Abstract
Mesenchymal stem cells (MSCs) are highly effective in the treatment of acute liver failure (ALF). The efficacy of MSCs is closely related to the inflammatory environment. Therefore, we investigated the functional changes of MSCs in response to interleukin-33 (IL-33) stimulation. The results showed that bone marrow mesenchymal stem cells (BMSCs) pretreated with IL-33 had increased CCR2 expression, targeted CCL2 in the injured liver tissue, and improved the migration ability. Under LPS stimulation, the NF-κB pathway of BMDM was activated, and its phenotype polarized to the M1-type, while BMSCs pretreated with IL-33 inhibited the NF-κB pathway and enhanced M2 macrophage polarization. The M2-type macrophages could further inhibit hepatocytes inflammation, reduce hepatocytes apoptosis, and promote hepatocytes repair. These results suggest that IL-33 can enhance the efficacy of BMSCs in ALF and provide a new strategy for cell therapy of liver diseases.
Collapse
Affiliation(s)
- Hui Yuan
- Department of Infectious Disease, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yuwen Li
- Department of Pediatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Zihao Kong
- Department of Gastroenterology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Linya Peng
- Department of Infectious Disease, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jiali Song
- Department of Infectious Disease, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xiaoxue Hou
- Department of Infectious Disease, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Wen Zhang
- Department of Infectious Disease, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Rui Liu
- Department of Infectious and Tropical Diseases, The Second Affiliated Hospital, NHC Key Laboratory of Tropical Disease Control, Hainan Medical University, Haikou, China
| | - Tiantong Feng
- Department of Infectious Disease, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Chuanlong Zhu
- Department of Infectious Disease, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Department of Infectious and Tropical Diseases, The Second Affiliated Hospital, NHC Key Laboratory of Tropical Disease Control, Hainan Medical University, Haikou, China
| |
Collapse
|
42
|
Yang Y, Wang J, Lin X, Zhang Z, Zhang M, Tang C, Kou X, Deng F. TNF-α-licensed exosome-integrated titaniumaccelerated T2D osseointegration by promoting autophagy-regulated M2 macrophage polarization. Biochem Biophys Res Commun 2024; 727:150316. [PMID: 38959732 DOI: 10.1016/j.bbrc.2024.150316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 06/18/2024] [Accepted: 06/24/2024] [Indexed: 07/05/2024]
Abstract
Type 2 diabetes (T2D) is on a notable rise worldwide, which leads to unfavorable outcomes during implant treatments. Surface modification of implants and exosome treatment have been utilized to enhance osseointegration. However, there has been insufficient approach to improve adverse osseointegration in T2D conditions. In this study, we successfully loaded TNF-α-treated mesenchymal stem cell (MSC)-derived exosomes onto micro/nano-network titanium (Ti) surfaces. TNF-α-licensed exosome-integrated titanium (TNF-exo-Ti) effectively enhanced M2 macrophage polarization in hyperglycemic conditions, with increased secretion of anti-inflammatory cytokines and decreased secretion of pro-inflammatory cytokines. In addition, TNF-exo-Ti pretreated macrophage further enhanced angiogenesis and osteogenesis of endothelial cells and bone marrow MSCs. More importantly, TNF-exo-Ti markedly promoted osseointegration in T2D mice. Mechanistically, TNF-exo-Ti activated macrophage autophagy to promote M2 polarization through inhibition of the PI3K/AKT/mTOR pathway, which could be abolished by PI3K agonist. Thus, this study established TNF-α-licensed exosome-immobilized titanium surfaces that could rectify macrophage immune states and accelerate osseointegration in T2D conditions.
Collapse
Affiliation(s)
- Yang Yang
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
| | - Jinyang Wang
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China; Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, South China Center of Craniofacial Stem Cell Research, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, 510055, China
| | - Xiaoxuan Lin
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
| | - Zhengchuan Zhang
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
| | - Manjin Zhang
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510055, China
| | - Cuizhu Tang
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
| | - Xiaoxing Kou
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China; Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, South China Center of Craniofacial Stem Cell Research, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, 510055, China.
| | - Feilong Deng
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China.
| |
Collapse
|
43
|
Cunha Silva L, Branco F, Cunha J, Vitorino C, Gomes C, Carrascal MA, Falcão A, Miguel Neves B, Teresa Cruz M. The potential of exosomes as a new therapeutic strategy for glioblastoma. Eur J Pharm Biopharm 2024; 203:114460. [PMID: 39218361 DOI: 10.1016/j.ejpb.2024.114460] [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: 10/03/2023] [Revised: 07/30/2024] [Accepted: 08/18/2024] [Indexed: 09/04/2024]
Abstract
Glioblastoma (GBM) stands for the most common and aggressive type of brain tumour in adults. It is highly invasive, which explains its short rate of survival. Little is known about its risk factors, and current therapy is still ineffective. Hence, efforts are underway to develop novel and effective treatment approaches against this type of cancer. Exosomes are being explored as a promising strategy for conveying and delivering therapeutic cargo to GBM cells. They can fuse with the GBM cell membrane and, consequently, serve as delivery systems in this context. Due to their nanoscale size, exosomes can cross the blood-brain barrier (BBB), which constitutes a significant hurdle to most chemotherapeutic drugs used against GBM. They can subsequently inhibit oncogenes, activate tumour suppressor genes, induce immune responses, and control cell growth. However, despite representing a promising tool for the treatment of GBM, further research and clinical studies regarding exosome biology, engineering, and clinical applications still need to be completed. Here, we sought to review the application of exosomes in the treatment of GBM through an in-depth analysis of the scientific and clinical studies on the entire process, from the isolation and purification of exosomes to their design and transformation into anti-oncogenic drug delivery systems. Surface modification of exosomes to enhance BBB penetration and GBM-cell targeting is also a topic of discussion.
Collapse
Affiliation(s)
- Leonor Cunha Silva
- Faculty of Pharmacy, FFUC, University of Coimbra, Coimbra 3000-548, Portugal
| | - Francisco Branco
- Faculty of Pharmacy, FFUC, University of Coimbra, Coimbra 3000-548, Portugal
| | - Joana Cunha
- Faculty of Pharmacy, FFUC, University of Coimbra, Coimbra 3000-548, Portugal
| | - Carla Vitorino
- Faculty of Pharmacy, FFUC, University of Coimbra, Coimbra 3000-548, Portugal; Coimbra Chemistry Centre, Institute of Molecular Sciences - IMS, Department of Chemistry, University of Coimbra, Coimbra 3004 535, Portugal
| | - Célia Gomes
- Coimbra Institute for Clinical and Biomedical Research, iCBR, Faculty of Medicine, University of Coimbra, Coimbra 3000-548, Portugal; Center for Innovation in Biomedicine and Biotechnology, CIBB, University of Coimbra, Coimbra 3000-504, Portugal
| | - Mylène A Carrascal
- Tecnimede Group, Sintra 2710-089, Portugal; Center for Neuroscience and Cell Biology, CNC, University of Coimbra, Coimbra 3004-504, Portugal
| | - Amílcar Falcão
- Faculty of Pharmacy, FFUC, University of Coimbra, Coimbra 3000-548, Portugal; Coimbra Institute for Biomedical Imaging and Translational Research, CIBIT, University of Coimbra, Coimbra 3000-548, Portugal
| | - Bruno Miguel Neves
- Department of Medical Sciences and Institute of Biomedicine, iBiMED, University of Aveiro, Aveiro 3810-193, Portugal
| | - Maria Teresa Cruz
- Faculty of Pharmacy, FFUC, University of Coimbra, Coimbra 3000-548, Portugal; Coimbra Institute for Clinical and Biomedical Research, iCBR, Faculty of Medicine, University of Coimbra, Coimbra 3000-548, Portugal; Center for Neuroscience and Cell Biology, CNC, University of Coimbra, Coimbra 3004-504, Portugal.
| |
Collapse
|
44
|
Zhong W, Xiong K, Li S, Li C. Macrophage polarization-related gene signature for risk stratification and prognosis of survival in gliomas. J Cell Mol Med 2024; 28:e70000. [PMID: 39448550 PMCID: PMC11502305 DOI: 10.1111/jcmm.70000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 01/12/2024] [Accepted: 02/09/2024] [Indexed: 10/26/2024] Open
Abstract
Macrophage polarization plays an essential role in tumour immune cell infiltration and tumour growth. In this study, we selected a series of genes distinguishing between M1 and M2 macrophages and explored their prognostic value in gliomas. A total of 170 genes were included in our study. The CGGA database was used as the training cohort and the TCGA database as the validation cohort. The biological processes and functions were identified by GO and KEGG analysis. Kaplan-Meier analysis was used to compare survival differences between groups. Importantly, we built a risk score model using Cox regression analysis based on the CGGA and verified it in the TCGA database and our sequencing data. Patients with gliomas in the high-risk group were associated with high pathologic grade, IDH WT status, MGMT promoter unmethylation, 1p19q non-codeletion and prone to have a poor outcome. GEPIA results revealed that CD300C, CNRIP1 and MYO1F are the most related genes of immune infiltrations. The differential expression of these genes between low-grade gliomas and glioblastomas was confirmed by q-RT-PCR. Macrophage polarization-related gene signatures can predict the malignancy and outcome of patients with gliomas and might act as a promising target for glioma immunotherapy in the future.
Collapse
Affiliation(s)
- Weiming Zhong
- Department of Neurosurgery, Xiangya HospitalCentral South UniversityChangshaPeople's Republic of China
- Hypothalamic‐Pituitary Research Center, Xiangya HospitalCentral South UniversityChangshaHunanPeople's Republic of China
| | - Kaifen Xiong
- Department of DermatologyShenzhen People’s Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology)ShenzhenGuangdongPeople's Republic of China
| | - Shuwang Li
- Department of NeurosurgeryThe Second People's Hospital of Hunan ProvinceChangshaPeople's Republic of China
| | - Chuntao Li
- Department of Neurosurgery, Xiangya HospitalCentral South UniversityChangshaPeople's Republic of China
- Hypothalamic‐Pituitary Research Center, Xiangya HospitalCentral South UniversityChangshaHunanPeople's Republic of China
| |
Collapse
|
45
|
Amiri V, Mirzaeian A, Noroozi-Aghideh A. Non-Mutational Changes of Autophagy Marker LC3A in Patients with Acute Myeloid Leukemia; Effect of DNA Methylation and Expression Level of LncRNA-GAS5 and miRNA-155-5p, A Case Control Study. Indian J Hematol Blood Transfus 2024; 40:621-628. [PMID: 39469184 PMCID: PMC11512980 DOI: 10.1007/s12288-024-01765-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 03/29/2024] [Indexed: 10/30/2024] Open
Abstract
Clinical translation of autophagy modulators is tied to thoroughly acquainted with the precise state of this process and its regulators in a particular cancer. LC3Av1 is a marker of autophagosome membrane that has been contributed with pathobiology of myriad of human cancers. In the present study, we examined the effect of promoter methylation and miR-155 and LncRNA-GAS5 (GAS5) expression levels on transcription of LC3Av1 in AML patients. The study included 60 patients with de novo AML and 20 subjects with normal bone marrow cellular composition. Methylation-Sensitive high resolution melting (MS-HRM) was performed for analysis of LC3Av1 CpG island methylation and reverse transcription-quantitative polymerase chain reaction (RT-qPCR) for assessing LC3Av1, GAS5 and miR-155 expression levels. There was a significant elevation in the expression level of miR-155 and repression of LC3Av1 in AML samples. We found that LC3Av1 downregulation was negatively associated with its CpG island hypermethylation and miR-155 expression. Aging leads to overexpression of LC3Av1. GAS5 neither was differently expressed in AML patients compared to control samples nor has been related to LC3Av1 expression. The present study revealed that epigenetic changes like DNA methylation and alteration of miR-155 have a pivotal role in repression of autophagy marker LC3Av1, which potentially could provide the important clues of prognostic and therapeutic targets. The optimal strategies for clinical implementation of autophagy in AML is yet to be fully achieved and deserve further studies. Supplementary Information The online version contains supplementary material available at 10.1007/s12288-024-01765-3.
Collapse
Affiliation(s)
- Vahid Amiri
- Department of Laboratory Sciences, School of Paramedicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Amin Mirzaeian
- HSCT Research Center, Shahid Beheshti University of Medical Science, Tehran, Iran
| | - Ali Noroozi-Aghideh
- HSCT Research Center, Shahid Beheshti University of Medical Science, Tehran, Iran
- Department of Hematology, Faculty of Paramedicine, Aja University of Medical Sciences, Tehran, Iran
| |
Collapse
|
46
|
Zhang L, Yan X, Wang Y, Wang Q, Yan H, Yan Y. Identification and validation of a novel robust glioblastoma prognosis model based on bioinformatics. Heliyon 2024; 10:e37374. [PMID: 39309926 PMCID: PMC11414505 DOI: 10.1016/j.heliyon.2024.e37374] [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: 05/28/2024] [Revised: 08/06/2024] [Accepted: 09/02/2024] [Indexed: 09/25/2024] Open
Abstract
Background Glioblastoma (GBM) is a very common primary malignant tumor of the central nervous system (CNS). Aging, macrophage, autophagy, and methylation related genes are hypothesized to be crucial to its pathogenesis. In this study, we aimed to explore the role of these genes in the prognosis of GBM. Methods The RNA sequence (RNA-seq) and clinical information were downloaded from The Cancer Genome Atlas database (TCGA) and the Chinese Glioma Genome Atlas database (CGGA). We performed univariate and least absolute shrinkage and selection operator (LASSO) multivariate Cox regression analysis to identify risk signatures related to overall survival (OS). We further developed a nomogram to predict individual outcomes. In addition, the immune microenvironment was analyzed by CIBERSORT. Results 256 differentially expressed genes (DEGs) were obtained based on aging, macrophage, autophagy, and methylation related genes between GBM samples and normal tissues in TCGA-GBM cohort. We identified five optimal risk signatures with prognostic values in TCGA-GBM cohort and established a prognostic risk score model. The validity of the model was verified in the CGGA cohort and Huanhu cohort. Finally, we constructed a nomogram for clinical application by combining age, O(6)-methylguanine-DNA methyltransferase (MGMT) promoter methylation status, and risk score. Activated NK cells and resting mast cells were highly expressed and memory B cells, plasma cells, resting NK cells, M1 macrophages, and neutrophils exhibited low expression in the high-risk score group. GBM patients with a low-risk score had a higher Tumor Immune Dysfunction and Exclusion (TIDE) score. The risk score of hot tumors was higher than that of the cold tumors. Additionally, 29 genes involved in glucose and lipid metabolism were highly expressed with a high-risk score. 31 metabolism-related pathways were significantly different between high-risk and low-risk groups. Conclusions We constructed and validated a novel prognostic model for GBM. Aging, macrophage, autophagy, and methylation related genes may serve as prognostic and therapeutic biomarkers. The model developed may assist in guiding treatment for GBM patients. Our research had great significance in accurately predicting the prognosis of GBM and may offer reference for immunotherapy decision for GBM patients.
Collapse
Affiliation(s)
- Le Zhang
- Clinical College of Neurology, Neurosurgery and Neurorehabilitation, Tianjin Medical University, Tianjin, 300070, China
- Department of Clinical Laboratory, Tianjin Huanhu Hospital, Tianjin, 300350, China
| | - Xiaoling Yan
- Department of Pathology, Tianjin Huanhu Hospital, Tianjin, 300350, China
| | - Yahong Wang
- Nero-oncology Center, Tianjin Huanhu Hospital, Tianjin, 300350, China
| | - Qin Wang
- Clinical College of Neurology, Neurosurgery and Neurorehabilitation, Tianjin Medical University, Tianjin, 300070, China
- Department of Clinical Laboratory, Tianjin Huanhu Hospital, Tianjin, 300350, China
| | - Hua Yan
- Clinical College of Neurology, Neurosurgery and Neurorehabilitation, Tianjin Medical University, Tianjin, 300070, China
- Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative Diseases, Tianjin Neurosurgical Institute, Tianjin Huanhu Hospital, Tianjin, 300350, China
| | - Yan Yan
- Clinical College of Neurology, Neurosurgery and Neurorehabilitation, Tianjin Medical University, Tianjin, 300070, China
- Medical Engineering and Translational Medicine Research Institute, Tianjin University of Medicine, Tianjin, 300072, China
- Department of Clinical Laboratory, Tianjin Huanhu Hospital, Tianjin, 300350, China
| |
Collapse
|
47
|
Kundu M, Das S, Dey A, Mandal M. Dual perspective on autophagy in glioma: Detangling the dichotomous mechanisms of signaling pathways for therapeutic insights. Biochim Biophys Acta Rev Cancer 2024; 1879:189168. [PMID: 39121913 DOI: 10.1016/j.bbcan.2024.189168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 06/25/2024] [Accepted: 08/05/2024] [Indexed: 08/12/2024]
Abstract
Autophagy is a normal physiological process that aids the recycling of cellular nutrients, assisting the cells to cope with stressed conditions. However, autophagy's effect on cancer, including glioma, is uncertain and involves complicated molecular mechanisms. Several contradictory reports indicate that autophagy may promote or suppress glioma growth and progression. Autophagy inhibitors potentiate the efficacy of chemotherapy or radiation therapy in glioma. Numerous compounds stimulate autophagy to cause glioma cell death. Autophagy is also involved in the therapeutic resistance of glioma. This review article aims to detangle the complicated molecular mechanism of autophagy to provide a better perception of the two-sided role of autophagy in glioma and its therapeutic implications. The protein and epigenetic modulators of the cytoprotective and cytotoxic role of autophagy are described in this article. Moreover, several signaling pathways are associated with autophagy and its effects on glioma. We have reviewed the molecular pathways and highlighted the signaling axis involved in cytoprotective and cytotoxic autophagy. Additionally, this article discusses the role of autophagy in therapeutic resistance, including glioma stem cell maintenance and tumor microenvironment regulation. It also summarizes several investigations on the anti-glioma effects of autophagy modulators to understand the associated mechanisms and provide insights regarding its therapeutic implications.
Collapse
Affiliation(s)
- Moumita Kundu
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, India; Center for Multidisciplinary Research & Innovations, Brainware University, Barasat, India; Department of Pharmaceutical Technology, Brainware University, Barasat, India.
| | - Subhayan Das
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, India; Department of Allied Health Sciences, Brainware University, Barasat, India
| | - Ankita Dey
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, India
| | - Mahitosh Mandal
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, India.
| |
Collapse
|
48
|
Lin Z, Hua G, Hu X. Lipid metabolism associated crosstalk: the bidirectional interaction between cancer cells and immune/stromal cells within the tumor microenvironment for prognostic insight. Cancer Cell Int 2024; 24:295. [PMID: 39174964 PMCID: PMC11342506 DOI: 10.1186/s12935-024-03481-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 08/13/2024] [Indexed: 08/24/2024] Open
Abstract
Cancer is closely related to lipid metabolism, with the tumor microenvironment (TME) containing numerous lipid metabolic interactions. Cancer cells can bidirectionally interact with immune and stromal cells, the major components of the TME. This interaction is primarily mediated by fatty acids (FAs), cholesterol, and phospholipids. These interactions can lead to various physiological changes, including immune suppression, cancer cell proliferation, dissemination, and anti-apoptotic effects on cancer cells. The physiological modulation resulting from this lipid metabolism-associated crosstalk between cancer cells and immune/stromal cells provides valuable insights into cancer prognosis. A comprehensive literature review was conducted to examine the function of the bidirectional lipid metabolism interactions between cancer cells and immune/stromal cells within the TME, particularly how these interactions influence cancer prognosis. A novel autophagy-extracellular vesicle (EV) pathway has been proposed as a mediator of lipid metabolism interactions between cancer cells and immune cells/stromal cells, impacting cancer prognosis. As a result, different forms of lipid metabolism interactions have been described as being linked to cancer prognosis, including those mediated by the autophagy-EV pathway. In conclusion, understanding the bidirectional lipid metabolism interactions between cancer cells and stromal/immune cells in the TME can help develop more advanced prognostic approaches for cancer patients.
Collapse
Affiliation(s)
- Zhongshu Lin
- Queen Mary College, Nanchang University, Nanchang, China
- School of Biological and Behavioural Science, Queen Mary University of London, London, UK
| | - Guanxiang Hua
- Queen Mary College, Nanchang University, Nanchang, China
- School of Biological and Behavioural Science, Queen Mary University of London, London, UK
| | - Xiaojuan Hu
- Queen Mary College, Nanchang University, Nanchang, China.
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, China.
| |
Collapse
|
49
|
Kim JH, Song JW, Kim YH, Kim HJ, Kim RH, Park YH, Nam HS, Kang DO, Yoo H, Park K, Kim JW. Multimodal Imaging-Assisted Intravascular Theranostic Photoactivation on Atherosclerotic Plaque. Circ Res 2024; 135:e114-e132. [PMID: 38989585 DOI: 10.1161/circresaha.123.323970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 07/02/2024] [Accepted: 07/03/2024] [Indexed: 07/12/2024]
Abstract
BACKGROUND Atherosclerosis is a chronic inflammatory disease causing a fatal plaque rupture, and its key aspect is a failure to resolve inflammation. We hypothesize that macrophage-targeted near-infrared fluorescence emitting photoactivation could simultaneously assess macrophage/lipid-rich plaques in vivo and facilitate inflammation resolution. METHODS We fabricated a Dectin-1-targeted photoactivatable theranostic agent through the chemical conjugation of the near-infrared fluorescence-emitting photosensitizer chlorin e6 and the Dectin-1 ligand laminarin (laminarin-chlorin e6 [LAM-Ce6]). Intravascular photoactivation by a customized fiber-based diffuser after administration of LAM-Ce6 effectively reduced inflammation in the targeted plaques of atherosclerotic rabbits in vivo as serially assessed by dual-modal optical coherence tomography-near-infrared fluorescence structural-molecular catheter imaging after 4 weeks. RESULTS The number of apoptotic macrophages peaked at 1 day after laser irradiation and then resolved until 4 weeks. Autophagy was strongly augmented 1 hour after the light therapy, with the formation of autophagolysosomes. LAM-Ce6 photoactivation increased the terminal deoxynucleotidyl transferase dUTP (deoxyuridine triphosphate) nick end labeling/RAM11 (rabbit monocyte/macrophage antibody)- and MerTK (c-Mer tyrosine kinase)-positive cells in the plaques, suggesting enhanced efferocytosis. In line with inflammation resolution, photoactivation reduced the plaque burden through fibrotic replacement via the TGF (transforming growth factor)-β/CTGF (connective tissue growth factor) pathway. CONCLUSIONS Optical coherence tomography-near-infrared fluorescence imaging-guided macrophage Dectin-1-targetable photoactivation could induce the transition of macrophage/lipid-rich plaques into collagen-rich lesions through autophagy-mediated inflammation resolution and TGF-β-dependent fibrotic replacement. This novel strategy offers a new opportunity for the catheter-based theranostic strategy.
Collapse
Affiliation(s)
- Jin Hyuk Kim
- BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Korea (J.H.K., J.W.K.)
- Multimodal Imaging and Theranostic Laboratory, Cardiovascular Center, Korea University Guro Hospital (J.H.K., J.W.S., H.J.K., R.H.K., Y.H.P., D.O.K., J.W.K.)
| | - Joon Woo Song
- Multimodal Imaging and Theranostic Laboratory, Cardiovascular Center, Korea University Guro Hospital (J.H.K., J.W.S., H.J.K., R.H.K., Y.H.P., D.O.K., J.W.K.)
| | - Yeon Hoon Kim
- Department of Mechanical Engineering, KAIST, Daejeon, Korea (Y.H.K., H.S.N., H.Y.)
| | - Hyun Jung Kim
- Multimodal Imaging and Theranostic Laboratory, Cardiovascular Center, Korea University Guro Hospital (J.H.K., J.W.S., H.J.K., R.H.K., Y.H.P., D.O.K., J.W.K.)
| | - Ryeong Hyun Kim
- Multimodal Imaging and Theranostic Laboratory, Cardiovascular Center, Korea University Guro Hospital (J.H.K., J.W.S., H.J.K., R.H.K., Y.H.P., D.O.K., J.W.K.)
| | - Ye Hee Park
- Multimodal Imaging and Theranostic Laboratory, Cardiovascular Center, Korea University Guro Hospital (J.H.K., J.W.S., H.J.K., R.H.K., Y.H.P., D.O.K., J.W.K.)
| | - Hyeong Soo Nam
- Department of Mechanical Engineering, KAIST, Daejeon, Korea (Y.H.K., H.S.N., H.Y.)
| | - Dong Oh Kang
- Multimodal Imaging and Theranostic Laboratory, Cardiovascular Center, Korea University Guro Hospital (J.H.K., J.W.S., H.J.K., R.H.K., Y.H.P., D.O.K., J.W.K.)
| | - Hongki Yoo
- Department of Mechanical Engineering, KAIST, Daejeon, Korea (Y.H.K., H.S.N., H.Y.)
| | - Kyeongsoon Park
- Department of Systems Biotechnology, Chung-Ang University, Anseong, Gyeonggi, Korea (K.P.)
| | - Jin Won Kim
- BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Korea (J.H.K., J.W.K.)
- Multimodal Imaging and Theranostic Laboratory, Cardiovascular Center, Korea University Guro Hospital (J.H.K., J.W.S., H.J.K., R.H.K., Y.H.P., D.O.K., J.W.K.)
| |
Collapse
|
50
|
Wang C, Xu S, Yang X. Hypoxia-Driven Changes in Tumor Microenvironment: Insights into Exosome-Mediated Cell Interactions. Int J Nanomedicine 2024; 19:8211-8236. [PMID: 39157736 PMCID: PMC11328847 DOI: 10.2147/ijn.s479533] [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: 05/23/2024] [Accepted: 08/06/2024] [Indexed: 08/20/2024] Open
Abstract
Hypoxia, as a prominent feature of the tumor microenvironment, has a profound impact on the multicomponent changes within this environment. Under hypoxic conditions, the malignant phenotype of tumor cells, the variety of cell types within the tumor microenvironment, as well as intercellular communication and material exchange, undergo complex alterations. These changes provide significant prospects for exploring the mechanisms of tumor development under different microenvironmental conditions and for devising therapeutic strategies. Exosomes secreted by tumor cells and stromal cells are integral components of the tumor microenvironment, serving as crucial mediators of intercellular communication and material exchange, and have consequently garnered increasing attention from researchers. This review focuses on the mechanisms by which hypoxic conditions promote the release of exosomes by tumor cells and alter their encapsulated contents. It also examines the effects of exosomes derived from tumor cells, immune cells, and other cell types under hypoxic conditions on the tumor microenvironment. Additionally, we summarize current research progress on the potential clinical applications of exosomes under hypoxic conditions and propose future research directions in this field.
Collapse
Affiliation(s)
- Churan Wang
- Dalian Medical University, Dalian, 116000, People’s Republic of China
| | - Shun Xu
- Department of Thoracic Surgery, The First Hospital of China Medical University, Shenyang, 110002, People’s Republic of China
| | - Xiao Yang
- Department of Thoracic Surgery, The First Hospital of China Medical University, Shenyang, 110002, People’s Republic of China
| |
Collapse
|