1
|
Li P, Guan D, Li S, Deng J, Zhang H, Liu X, Chen X, Xu Z, Wang H, Ren F. Exosomes derived from myelodysplastic syndromes cells induce IL-1β production from macrophages to promote disease progress. Cytokine 2025; 190:156924. [PMID: 40147377 DOI: 10.1016/j.cyto.2025.156924] [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: 12/19/2024] [Revised: 03/10/2025] [Accepted: 03/18/2025] [Indexed: 03/29/2025]
Abstract
BACKGROUND Exosomes are extracellular vesicles with a membrane structure that play important roles in intercellular communication, material transport and cellular immunity.Our previous study found that exosomes can affect the biological functions of MDS cell lines, but the mechanism of action has not been elucidated.Macrophages are one of the major innate immune cells that produce a variety of inflammatory cytokines and perform multiple biological functions in the tumor microenvironment (TME).The role of tumor cell-derived exosomes on macrophages and in the progression of MDS is rarely reported,therefore, the aim of our study was to investigate the effect of exosomes on macrophages and the effect of cytokines secreted by macrophages on MDS cells, with a view to exploring the role and mechanisms of exosomes and macrophages in the progression of MDS. METHODS Changes in cytokine content in peripheral blood of MDS patients were detected. The cytokine concentration in the growth environment of MDS cell lines was changed to observe the changes in the biological functions of MDS cell lines.After induction of human monocyte cell line (THP-1) into THP-1-Mφ macrophages with Phorbol 12-myristate 13-acetate (PMA), the macrophages (Mφ) were then co-cultured with MDS cell line exosomes extracted by ultrafiltration with THP-1-Mφ to observe macrophage (Mφ) differentiation.Flow cytometry was used to detect the changes in cytokine content released by macrophages before and after the addition of exosome inhibitors, and the changes in the biological function of MDS cell lines during this process.Gene and protein levels of significantly changed cytokine-related signaling pathways were detected using Q-PCR and WB. RESULTS IL-1β levels were significantly higher in the peripheral blood of MDS patients compared to controls.The exosomes extracted by ultrafiltration can be taken up by macrophages, which can promote the release of IL-1β from THP-1-Mφ cells, and promote the proliferation, apoptosis and migration ability of MDS cell lines.Exosomes stimulate macrophages to produce IL-1β and promote MDS disease progression through the MER/ERK pathway.
Collapse
Affiliation(s)
- Peichun Li
- Shanxi Medical University, 56 Xinjian South Road, Taiyuan, Shanxi Province, China
| | - Dongmei Guan
- Shanxi Medical University, 56 Xinjian South Road, Taiyuan, Shanxi Province, China
| | - Shuo Li
- Shanxi Medical University, 56 Xinjian South Road, Taiyuan, Shanxi Province, China
| | - Ju Deng
- Shanxi Medical University, 56 Xinjian South Road, Taiyuan, Shanxi Province, China
| | - HongYu Zhang
- Shanxi Medical University, 56 Xinjian South Road, Taiyuan, Shanxi Province, China
| | - Xiaoli Liu
- Shanxi Medical University, 56 Xinjian South Road, Taiyuan, Shanxi Province, China
| | - Xiuhua Chen
- Laboratory of Hematology, Second Hospital of Shanxi Medical University, Taiyuan, Shanxi Province, China; The Key Laboratory of Molecular Diagnosis and Treatment of Hematological Diseases of Shanxi Province, 382 Wuyi Road, Taiyuan, Shanxi Province, China
| | - Zhifang Xu
- Laboratory of Hematology, Second Hospital of Shanxi Medical University, Taiyuan, Shanxi Province, China; The Key Laboratory of Molecular Diagnosis and Treatment of Hematological Diseases of Shanxi Province, 382 Wuyi Road, Taiyuan, Shanxi Province, China
| | - Hongwei Wang
- Shanxi Medical University, 56 Xinjian South Road, Taiyuan, Shanxi Province, China; Laboratory of Hematology, Second Hospital of Shanxi Medical University, Taiyuan, Shanxi Province, China; The Key Laboratory of Molecular Diagnosis and Treatment of Hematological Diseases of Shanxi Province, 382 Wuyi Road, Taiyuan, Shanxi Province, China; Shanxi University of Chinese Medicine,No. 121, University Street, Yuci District, Jinzhong, City, Shanxi Province, China.
| | - Fanggang Ren
- Laboratory of Hematology, Second Hospital of Shanxi Medical University, Taiyuan, Shanxi Province, China; The Key Laboratory of Molecular Diagnosis and Treatment of Hematological Diseases of Shanxi Province, 382 Wuyi Road, Taiyuan, Shanxi Province, China.
| |
Collapse
|
2
|
Weiser T, Hoch CC, Petry J, Shoykhet M, Schmidl B, Yazdi M, Hachani K, Mergner J, Theodoraki MN, Azimzadeh O, Multhoff G, Bashiri Dezfouli A, Wollenberg B. Head and neck squamous cell carcinoma-derived extracellular vesicles mediate Ca²⁺-dependent platelet activation and aggregation through tissue factor. Cell Commun Signal 2025; 23:210. [PMID: 40312345 PMCID: PMC12044835 DOI: 10.1186/s12964-025-02215-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Accepted: 04/25/2025] [Indexed: 05/03/2025] Open
Abstract
BACKGROUND Head and neck squamous cell carcinoma (HNSCC) is an aggressive malignancy, characterized by poor clinical outcomes, primarily driven by high rate of locoregional recurrence and metastasis. Extensive heterogeneity among the tumor cells as well as modulation of a highly immunosuppressive tumor microenvironment shape cancer progression. Shedding of extracellular vesicles (EVs) derived from tumor cells is a critical mediator of the disease initiating horizontal transfer of tumor components into platelets. This triggers platelet activation and thromboinflammation fueling tumor progression through multiple mechanisms. METHODS HNSCC-derived EVs isolated from HNSCC cell lines (SAS, UD-SCC 5) using size exclusion chromatography and characterized via flow cytometry, electron microscopy, nanoparticle tracking analysis and Western blotting, were used to induce platelet activation and aggregation, measured by aggregometry, flow cytometry, as well as the release of chemokines and Adenosine triphosphate, which were quantified using enzyme-linked immunosorbent assays (ELISA). Mechanistic investigations included inhibitor assays, thrombin activity measurements, and proteomic analyses. RESULTS We could show that EVs do not activate platelets through the FcγRIIa-IgG axis but platelet activation and aggregation is induced in a calcium-dependent manner, primarily mediated by EV-associated tissue factor. Proteomic analysis confirmed the presence of tissue factor in these vesicles, implicating its involvement in initiating the coagulation cascade, that leads to platelet activation and aggregation. This process was characterized by delayed aggregation kinetics and relied on thrombin activation, as the inhibition of thrombin and its receptors reduced platelet aggregation. HNSCC-derived EVs are pivotal in establishing a prothrombotic environment by promoting platelet activation and aggregation through tissue factor-dependent thrombin generation. CONCLUSION These findings indicate a therapeutic potential of targeting EV-mediated pathways as a therapeutic approach to alleviate thrombotic complications in HNSCC patients. Subsequent animal studies will be crucial to validate and extend these observations, providing deeper insight into their clinical implications.
Collapse
Affiliation(s)
- Tobias Weiser
- Department of Otolaryngology, Head and Neck Surgery, TUM School of Medicine and Health, Technical University of Munich (TUM), Munich, Germany
| | - Cosima C Hoch
- Department of Otolaryngology, Head and Neck Surgery, TUM School of Medicine and Health, Technical University of Munich (TUM), Munich, Germany
| | - Julie Petry
- Department of Otolaryngology, Head and Neck Surgery, TUM School of Medicine and Health, Technical University of Munich (TUM), Munich, Germany
| | - Maria Shoykhet
- Department of Otolaryngology, Head and Neck Surgery, TUM School of Medicine and Health, Technical University of Munich (TUM), Munich, Germany
| | - Benedikt Schmidl
- Department of Otolaryngology, Head and Neck Surgery, TUM School of Medicine and Health, Technical University of Munich (TUM), Munich, Germany
| | - Mina Yazdi
- Pharmaceutical Biotechnology, Department of Pharmacy, Ludwig-Maximilians-Universität (LMU), Munich, Germany
| | - Khouloud Hachani
- Department of Otolaryngology, Head and Neck Surgery, TUM School of Medicine and Health, Technical University of Munich (TUM), Munich, Germany
| | - Julia Mergner
- Bavarian Center for Biomolecular Mass Spectrometry at Klinikum rechts der Isar (BayBioMS@MRI), TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Marie-Nicole Theodoraki
- Department of Otolaryngology, Head and Neck Surgery, TUM School of Medicine and Health, Technical University of Munich (TUM), Munich, Germany
| | - Omid Azimzadeh
- Section Radiation Biology, Federal Office for Radiation Protection (BfS), Neuherberg, Germany
| | - Gabriele Multhoff
- Central Institute for Translational Cancer Research, Department of Radiation Oncology, TUM School of Medicine and Health, Technical University of Munich (TranslaTUM), Technical University of Munich, Munich, Germany
| | - Ali Bashiri Dezfouli
- Department of Otolaryngology, Head and Neck Surgery, TUM School of Medicine and Health, Technical University of Munich (TUM), Munich, Germany
- Central Institute for Translational Cancer Research, Department of Radiation Oncology, TUM School of Medicine and Health, Technical University of Munich (TranslaTUM), Technical University of Munich, Munich, Germany
| | - Barbara Wollenberg
- Department of Otolaryngology, Head and Neck Surgery, TUM School of Medicine and Health, Technical University of Munich (TUM), Munich, Germany.
| |
Collapse
|
3
|
Wang Q, Sun J, Jiang H, Yu M. Emerging roles of extracellular vesicles in oral and maxillofacial areas. Int J Oral Sci 2025; 17:11. [PMID: 39900916 PMCID: PMC11791077 DOI: 10.1038/s41368-024-00341-9] [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: 05/03/2024] [Revised: 12/02/2024] [Accepted: 12/03/2024] [Indexed: 02/05/2025] Open
Abstract
The oral and maxillofacial region is a highly complex area composed of multiple tissue types and bears various critical functions of the human body. Diseases in this region pose significant diagnostic and management challenges; therefore, exploring new strategies for early diagnosis, targeted treatment, and tissue reconstruction is key to improving patient prognosis and quality of life. Extracellular vesicles are a group of heterogeneous lipid-bilayer membrane structures secreted by most cell types, including exosomes, microvesicles, and apoptotic bodies. Present in various body fluids and tissues, they act as messengers via the transfer of nucleic acids, proteins, and metabolites to recipient cells. To date, studies have revealed the different roles of extracellular vesicles in physiological or pathological processes, as well as applications in disease diagnosis, prognosis, and treatment. The importance and tissue specificity of the dental and maxillofacial tissues indicate that extracellular vesicles derived from this region are promising for further research. This paper reviews the published data on extracellular vesicles derived from cells, body fluids, and tissues in oral and maxillofacial regions, summarizes the latest advances in extracellular vesicles from extensive sources, and concludes with a focus on the current research progress and application prospects of engineered exosomes in oral science.
Collapse
Affiliation(s)
- Qianting Wang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Diseases of the Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, China
| | - Jiayu Sun
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Diseases of the Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, China
| | - Haci Jiang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Diseases of the Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, China
| | - Mengfei Yu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Diseases of the Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, China.
| |
Collapse
|
4
|
Wang X, Xiang Z, Zhang Y, Tu CR, Huang C, Chung Y, Zhang W, Wang M, Liu Y, Tu W. CD25 downregulation by tumor exosomal microRNA-15a promotes interleukin-17-producing γδ-T-cells-mediated radioresistance in nasopharyngeal carcinoma. MedComm (Beijing) 2025; 6:e70078. [PMID: 39901895 PMCID: PMC11788015 DOI: 10.1002/mco2.70078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 12/04/2024] [Accepted: 01/02/2025] [Indexed: 02/05/2025] Open
Abstract
Interleukin (IL)-17-producing γδ-T cells (γδT-17) are a major source of IL-17 within the tumor microenvironment and have been shown to influence tumor development and therapy outcomes in various cancers. However, the role and presence of γδT-17 cells in nasopharyngeal carcinoma (NPC) remain poorly understood. It is also unclear how these cells might affect radiotherapy, the primary treatment for NPC patients. In this study, we discovered that NPC tumor tissues were rich in γδT-17 cells. Exosomes released from NPC cells (NPC-Exos) could direct γδ-T cells to differentiate into γδT-17 cells. These NPC-Exos-induced γδT-17 cells were found to enhance radioresistance in NPC, both in vitro and in vivo. Blocking IL-17 secreted by NPC-Exos-induced γδT-17 cells restored NPC cell sensitivity to radiation and elevated radiation-induced cell death. Mechanistic studies revealed that NPC-Exos not only increased the release of IL-17-promoting cytokines IL-1β, IL-6, and IL-23 from dendritic cells, but also suppressed CD25/IL-2 signaling in γδ-T cells, facilitating γδT-17 differentiation. The suppression of CD25/IL-2 signaling was driven by microRNA-15a (miR-15a) carried by NPC exosomes. Furthermore, miR-15a inhibitors were able to prevent γδT-17 induction by NPC-Exos. Our findings reveal a novel immunoregulatory role of NPC-Exos and offer potential strategies to combat NPC radioresistance.
Collapse
Affiliation(s)
- Xiwei Wang
- Department of Paediatrics and Adolescent MedicineLi Ka Shing Faculty of MedicineUniversity of Hong KongHong Kong SARChina
- CAS Key Laboratory of Quantitative Engineering BiologyShenzhen Institute of Synthetic BiologyShenzhen Institute of Advanced TechnologyChinese Academy of SciencesShenzhenChina
| | - Zheng Xiang
- Department of Paediatrics and Adolescent MedicineLi Ka Shing Faculty of MedicineUniversity of Hong KongHong Kong SARChina
- Department of Microbiology and ImmunologyHealth Science Center (School of Medicine)Jinan UniversityJinanChina
| | - Yanmei Zhang
- Department of Paediatrics and Adolescent MedicineLi Ka Shing Faculty of MedicineUniversity of Hong KongHong Kong SARChina
- CAS Key Laboratory of Quantitative Engineering BiologyShenzhen Institute of Synthetic BiologyShenzhen Institute of Advanced TechnologyChinese Academy of SciencesShenzhenChina
| | - Chloe Ran Tu
- Department of data sciencesDana‐Farber Cancer InstituteHarvard UniversityBostonMassachusettsUSA
| | - Chunyu Huang
- Shenzhen Key Laboratory for Reproductive Immunology of Peri‐implantationShenzhen Zhongshan Institute for Reproduction and GeneticsShenzhen Zhongshan Obstetrics & Gynecology Hospital (formerly Shenzhen Zhongshan Urology Hospital)ShenzhenChina
| | - Yuet Chung
- Department of Paediatrics and Adolescent MedicineLi Ka Shing Faculty of MedicineUniversity of Hong KongHong Kong SARChina
| | - Wenyue Zhang
- Department of Paediatrics and Adolescent MedicineLi Ka Shing Faculty of MedicineUniversity of Hong KongHong Kong SARChina
| | - Manni Wang
- Department of Paediatrics and Adolescent MedicineLi Ka Shing Faculty of MedicineUniversity of Hong KongHong Kong SARChina
| | - Yinping Liu
- Department of Paediatrics and Adolescent MedicineLi Ka Shing Faculty of MedicineUniversity of Hong KongHong Kong SARChina
| | - Wenwei Tu
- Department of Paediatrics and Adolescent MedicineLi Ka Shing Faculty of MedicineUniversity of Hong KongHong Kong SARChina
- CAS Key Laboratory of Quantitative Engineering BiologyShenzhen Institute of Synthetic BiologyShenzhen Institute of Advanced TechnologyChinese Academy of SciencesShenzhenChina
| |
Collapse
|
5
|
Abousaad S, Ahmed F, Abouzeid A, Adhiambo C, Ongeri E. Meprin β activity modulates cellular proliferation via trans-signaling IL-6-mediated AKT/ERK pathway in IR-induced kidney injury. RESEARCH SQUARE 2025:rs.3.rs-5901359. [PMID: 39975921 PMCID: PMC11838750 DOI: 10.21203/rs.3.rs-5901359/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Inflammation plays a central role in the progression of kidney injury induced by ischemia/reperfusion (IR). Meprin metalloproteinases have been implicated in the pathophysiology of IR-induced kidney injury. Existing data from in vitro and in vivo studies show that meprins modulate interleukin-6 (IL-6)-mediated inflammation via proteolytic processing of IL-6 and its receptor. IL-6 trans-signaling induces proliferation through either MAPK/ERK or PI3K/AKT pathway or in crosstalk with AKT/ERK. We previously showed that meprin β modulates cellular survival (BCL-2) through IL-6/JAK/STAT signaling pathway in IR-induced kidney injury. However, it's not known how meprin β modulation of the IL-6 signaling pathway impacts the cellular proliferation in IR-induced acute kidney injury. The goal of the current study was to determine how meprin β modulation of the IL-6 signaling pathway impacts downstream cellular proliferation in IR-induced kidney injury. We used the unilateral IR as a model of renal inflammation in wild-type (WT) and meprin β knockout (βKO) mice, with the contralateral kidneys serving as controls. The mice were sacrificed at 96 h post-IR, and kidney tissue processed for evaluation by RT-PCR and immunohistochemistry. Statistical analysis utilized two-way ANOVA. RT-PCR data showed a significant increase in mRNA levels for IL-6 and proliferating cell nuclear antigen (PCNA) in WT and βKO mice at 96 h-post IR when compared to WT control kidneys. However, the baseline mRNA levels for PCNA were significantly higher in βKO when compared to WT kidneys. Immunohistochemical data showed significant increases in IL-6, PCNA, p-AKT and p-ERK in select tubules in both genotypes at 96 h post-IR when compared to control kidneys for each genotype. Data from immunofluorescence counterstaining of kidney tissues revealed that at 96 hours post-IR, IL-6, PCNA, p-AKT, and p-ERK were primarily expressed in meprin β-expressing proximal tubules (PTs), where meprins are abundantly present. However, high levels of IL-6 were also present in the lumen of PTs and DTs from WT and βKO kidneys at 96 h post-IR, suggesting increased release/shedding into filtrate and subsequently into urine. In conclusion, this study highlights the role of meprin β activity in regulating cellular proliferation through PCNA regulation, driven by the IL-6-mediated AKT/ERK signaling pathway during the recovery phase following IR-induced kidney injury.
Collapse
Affiliation(s)
| | - Faihaa Ahmed
- the College of Veterinary Medicine, North Carolina State University
| | | | | | | |
Collapse
|
6
|
Liang C, Kan J, Wang J, Lu W, Mo X, Zhang B. Nasopharyngeal carcinoma-associated inflammatory cytokines: ongoing biomarkers. Front Immunol 2024; 15:1448012. [PMID: 39483474 PMCID: PMC11524805 DOI: 10.3389/fimmu.2024.1448012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 10/02/2024] [Indexed: 11/03/2024] Open
Abstract
Nasopharyngeal carcinoma (NPC) is a neoplasm related to inflammation; the expression of cytokines, such as CCL3, CCL4, CCL20, IL-1α, IL-1β, IL-6, IL-8, and IL-10, among others, is presumed to be associated with NPC occurrence and development. Therefore, the circulating levels of these cytokines may be potential biomarkers for assessing tumor aggressiveness, exploring cellular interactions, and monitoring tumor therapeutic responses. Numerous scholars have comprehensively explored the putative mechanisms through which these inflammatory factors affect NPC progression and therapeutic responses. Moreover, investigations have focused on elucidating the correlation between the systemic levels of these cytokines and the incidence and prognosis of NPC. This comprehensive review aims to delineate the advancements in research concerning the relationship between inflammatory factors and NPC while considering their prospective roles as novel prognostic and predictive biomarkers in the context of NPC.
Collapse
Affiliation(s)
- Chuwen Liang
- TCM&VIP Inpatient Department, Sun Yat-sen University Cancer Center, Guangzhou, China
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Jun Kan
- TCM&VIP Inpatient Department, Sun Yat-sen University Cancer Center, Guangzhou, China
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Jingli Wang
- TCM&VIP Inpatient Department, Sun Yat-sen University Cancer Center, Guangzhou, China
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Wei Lu
- TCM&VIP Inpatient Department, Sun Yat-sen University Cancer Center, Guangzhou, China
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xiaoyan Mo
- TCM&VIP Inpatient Department, Sun Yat-sen University Cancer Center, Guangzhou, China
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Bei Zhang
- TCM&VIP Inpatient Department, Sun Yat-sen University Cancer Center, Guangzhou, China
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| |
Collapse
|
7
|
Zhu L, Duan W, Peng L, Shan X, Liu Y, Huang Z, Da Y, Han Y. A novel proteomic prognostic signature characterizes the immune landscape and predicts nasopharyngeal carcinoma prognosis. Heliyon 2024; 10:e37897. [PMID: 39386833 PMCID: PMC11462186 DOI: 10.1016/j.heliyon.2024.e37897] [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: 05/24/2024] [Revised: 09/12/2024] [Accepted: 09/12/2024] [Indexed: 10/12/2024] Open
Abstract
Background Nasopharyngeal carcinoma (NPC) is a highly diverse and aggressive cancer type, leading to varying prognoses and responses to immunotherapy. This study aims to develop a protein-based signature that provides new insights into assessing the prognosis and immunotherapeutic response in NPC patients. Methods and Results We obtained transcriptomic and proteomic data for NPC from TCGA and CPTAC databases, respectively. Differentially expressed proteins with prognostic significance were identified using limma combined with uniCox analysis. A prognostic protein signature was created utilizing the LASSO algorithm. Receiver operating characteristic (ROC) curve analysis along with Kaplan-Meier survival analysis was conducted to assess the predictive accuracy of this signature. To evaluate immune infiltration levels among patients categorized by high or low risk scores (RPscores), we employed ssGSEA and ESTIMATE methods, while TIDE was used to forecast responses to immunotherapy. Our research pinpointed four critical prognostic proteins: CdSTA, AGR3, DUSP14, and LRRC17, allowing us to compute risk scores (RPscores). Kaplan-Meier curves demonstrated that individuals in the low-risk category exhibited better survival rates. Furthermore, RPscore effectively predicted overall survival across both training and testing cohorts. The ssGSEA results indicated that RPscore is linked with an immune-suppressive microenvironment correlating with diminished immune responses. Notably, DUSP14 showed significant upregulation in NPC cases; its role in promoting cell invasion and metastasis was confirmed through in vitro studies. Conclusion We have established a robust protein-related signature capable of accurately forecasting prognosis as well as immunotherapy outcomes for NPC patients. Moreover, DUSP14 emerged as a potential therapeutic target due to its strong association with patient prognosis in nasopharyngeal carcinoma.
Collapse
Affiliation(s)
- Lixin Zhu
- Department of Otolaryngology, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai, 201318, China
| | - Wenliang Duan
- Department of Otolaryngology, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai, 201318, China
| | - Lijing Peng
- Department of Otolaryngology, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai, 201318, China
| | - Xinxin Shan
- Department of Otolaryngology, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai, 201318, China
| | - Yuan Liu
- Department of Otolaryngology, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai, 201318, China
| | - Zhenke Huang
- Department of Otolaryngology, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai, 201318, China
| | - Yunxiang Da
- Department of Otolaryngology, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai, 201318, China
| | - Yanyan Han
- Department of Otolaryngology, Shanghai Punan Hospital, Shanghai, 200120, China
| |
Collapse
|
8
|
Xie HJ, Jiang MJ, Jiang K, Tang LQ, Chen QY, Yang AK, Mai HQ. Communication between cancer cell subtypes by exosomes contributes to nasopharyngeal carcinoma metastasis and poor prognosis. PRECISION CLINICAL MEDICINE 2024; 7:pbae018. [PMID: 39347440 PMCID: PMC11427951 DOI: 10.1093/pcmedi/pbae018] [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: 05/05/2024] [Accepted: 08/25/2024] [Indexed: 10/01/2024] Open
Abstract
Background Intratumor heterogeneity is common in cancers, with different cell subtypes supporting each other to become more malignant. Nasopharyngeal carcinoma (NPC), a highly metastatic cancer, shows significant heterogeneity among its cells. This study investigates how NPC cell subtypes with varying metastatic potentials influence each other through exosome-transmitted molecules. Methods Exosomes were purified and characterized. MicroRNA expression was analyzed via sequencing and qRT-PCR. The effects of miR-30a-5p on migration, invasion, and metastasis were evaluated in vitro and in vivo. Its impact on desmoglein glycoprotein (DSG2) was assessed using dual-luciferase assays and Western blotting. Immunohistochemistry (IHC) and statistical models linked miR-30a-5p/DSG2 levels to patient prognosis. Results Different NPC cell subtypes transmit metastatic potential via exosomes. High-metastatic cells enhance the migration, invasion, and metastasis of low-metastatic cells through exosome-transmitted miR-30a-5p. Plasma levels of exosomal miR-30a-5p are reliable indicators of NPC prognosis. miR-30a-5p may promote metastasis by targeting DSG2 and modulating Wnt signaling. Plasma exosomal miR-30a-5p inversely correlates with DSG2 levels, predicting patient outcomes. Conclusion High-metastatic NPC cells can increase the metastatic potential of low-metastatic cells through exosome-transmitted miR-30a-5p, which is a valuable prognostic marker assessable via liquid biopsy.
Collapse
Affiliation(s)
- Hao-Jun Xie
- Department of Head and Neck, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Ming-Jie Jiang
- Department of Head and Neck, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Ke Jiang
- Department of Head and Neck, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Lin-Quan Tang
- Departcment of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Qiu-Yan Chen
- Departcment of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - An-Kui Yang
- Department of Head and Neck, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Hai-Qiang Mai
- Departcment of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| |
Collapse
|
9
|
Zhu Y, Lu Y, Xu C, Huang Y, Yu Z, Wang T, Mao L, Liao X, Li S, Zhang W, Zhou F, Liu K, Zhang Y, Yang W, Min S, Deng Y, Wang Z, Fan X, Nie G, Xie X, Li Z. TMEM52B Isoforms P18 and P20 Differentially Promote the Oncogenesis and Metastasis of Nasopharyngeal Carcinoma. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2402457. [PMID: 38940427 PMCID: PMC11434218 DOI: 10.1002/advs.202402457] [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: 03/07/2024] [Revised: 05/29/2024] [Indexed: 06/29/2024]
Abstract
Transmembrane protein 52B (TMEM52B), a newly identified tumor-related gene, has been reported to regulate various tumors, yet its role in nasopharyngeal carcinoma (NPC) remains unclear. Transcriptomic analysis of NPC cell lines reveals frequent overexpression of TMEM52B, and immunohistochemical results show that TMEM52B is associated with advanced tumor stage, recurrence, and decreased survival time. Depleting TMEM52B inhibits the proliferation, migration, invasion, and oncogenesis of NPC cells in vivo. TMEM52B encodes two isoforms, TMEM52B-P18 and TMEM52B-P20, differing in their N-terminals. While both isoforms exhibit similar pro-oncogenic roles and contribute to drug resistance in NPC, TMEM52B-P20 differentially promotes metastasis. This functional discrepancy may be attributed to their distinct subcellular localization; TMEM52B-P18 is confined to the cytoplasm, while TMEM52B-P20 is found both at the cell membrane and in the cytoplasm. Mechanistically, cytoplasmic TMEM52B enhances AKT phosphorylation by interacting with phosphoglycerate kinase 1 (PGK1), fostering NPC growth and metastasis. Meanwhile, membrane-localized TMEM52B-P20 promotes E-cadherin ubiquitination and degradation by facilitating its interaction with the E3 ubiquitin ligase NEDD4, further driving NPC metastasis. In conclusion, the TMEM52B-P18 and TMEM52B-P20 isoforms promote the metastasis of NPC cells through different mechanisms. Drugs targeting these TMEM52B isoforms may offer therapeutic benefits to cancer patients with varying degrees of metastasis.
Collapse
Affiliation(s)
- Yuqi Zhu
- Guangdong Provincial Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Shenzhen Key Laboratory of Genitourinary Tumor, Department of Urology, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, 518000, China
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen, 518060, China
- Medical Research Center, The Affiliated Yue Bei People's Hospital, Shantou University Medical College, Shaoguan, 512025, China
| | - Yanxin Lu
- Guangdong Provincial Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Shenzhen Key Laboratory of Genitourinary Tumor, Department of Urology, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, 518000, China
- Basic Medical Science Department, Zhuhai Campus of Zunyi Medical University, Zhuhai, 519041, China
| | - Chunhua Xu
- Guangdong Provincial Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Shenzhen Key Laboratory of Genitourinary Tumor, Department of Urology, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, 518000, China
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen, 518060, China
| | - Yuqian Huang
- Guangdong Provincial Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Shenzhen Key Laboratory of Genitourinary Tumor, Department of Urology, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, 518000, China
| | - Ziyi Yu
- Guangdong Provincial Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Shenzhen Key Laboratory of Genitourinary Tumor, Department of Urology, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, 518000, China
| | - Tongyu Wang
- Guangdong Provincial Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Shenzhen Key Laboratory of Genitourinary Tumor, Department of Urology, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, 518000, China
| | - Longyi Mao
- Guangdong Provincial Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Shenzhen Key Laboratory of Genitourinary Tumor, Department of Urology, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, 518000, China
| | - Ximian Liao
- Guangdong Provincial Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Shenzhen Key Laboratory of Genitourinary Tumor, Department of Urology, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, 518000, China
| | - Shi Li
- Guangdong Provincial Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Shenzhen Key Laboratory of Genitourinary Tumor, Department of Urology, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, 518000, China
| | - Wanqing Zhang
- Guangdong Provincial Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Shenzhen Key Laboratory of Genitourinary Tumor, Department of Urology, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, 518000, China
| | - Feng Zhou
- Oncology Department, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518060, China
| | - Kaiqing Liu
- Guangdong Provincial Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Shenzhen Key Laboratory of Genitourinary Tumor, Department of Urology, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, 518000, China
| | - Yu Zhang
- Guangdong Provincial Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Shenzhen Key Laboratory of Genitourinary Tumor, Department of Urology, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, 518000, China
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen, 518060, China
| | - Wei Yang
- Guangdong Provincial Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Shenzhen Key Laboratory of Genitourinary Tumor, Department of Urology, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, 518000, China
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen, 518060, China
| | - Shasha Min
- Guangdong Provincial Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Shenzhen Key Laboratory of Genitourinary Tumor, Department of Urology, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, 518000, China
- Basic Medical Science Department, Zhuhai Campus of Zunyi Medical University, Zhuhai, 519041, China
| | - Yaqin Deng
- Guangdong Provincial Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Shenzhen Key Laboratory of Genitourinary Tumor, Department of Urology, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, 518000, China
| | - Zaixing Wang
- Institute of Otorhinolaryngology and Shenzhen Key of Otorhinolaryngology, Longgang Otorhinolaryngology Hospital, Shenzhen, 518172, China
| | - Xiaoqin Fan
- The Bio-bank of Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong, 518000, China
| | - Guohui Nie
- The Bio-bank of Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong, 518000, China
| | - Xina Xie
- Guangdong Provincial Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Shenzhen Key Laboratory of Genitourinary Tumor, Department of Urology, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, 518000, China
| | - Zesong Li
- Guangdong Provincial Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Shenzhen Key Laboratory of Genitourinary Tumor, Department of Urology, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, 518000, China
- Medical Research Center, The Affiliated Yue Bei People's Hospital, Shantou University Medical College, Shaoguan, 512025, China
- Basic Medical Science Department, Zhuhai Campus of Zunyi Medical University, Zhuhai, 519041, China
- Institute of Basic Medicine and Forensic Medicine, North Sichuan Medical College, Nanchong, Sichuan, 637199, China
| |
Collapse
|
10
|
Chak PT, Kam NW, Choi TH, Dai W, Kwong DLW. Unfolding the Complexity of Exosome-Cellular Interactions on Tumour Immunity and Their Clinical Prospects in Nasopharyngeal Carcinoma. Cancers (Basel) 2024; 16:919. [PMID: 38473281 DOI: 10.3390/cancers16050919] [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: 01/26/2024] [Revised: 02/19/2024] [Accepted: 02/23/2024] [Indexed: 03/14/2024] Open
Abstract
Nasopharyngeal carcinoma (NPC) is an epithelial malignancy situated in the posterolateral nasopharynx. NPC poses grave concerns in Southeast Asia due to its late diagnosis. Together with resistance to standard treatment combining chemo- and radiotherapy, NPC presents high metastatic rates and common recurrence. Despite advancements in immune-checkpoint inhibitors (ICIs) and cytotoxic-T-lymphocytes (CTLs)-based cellular therapy, the exhaustive T cell profile and other signs of immunosuppression within the NPC tumour microenvironment (TME) remain as concerns to immunotherapy response. Exosomes, extracellular vesicles of 30-150 nm in diameter, are increasingly studied and linked to tumourigenesis in oncology. These bilipid-membrane-bound vesicles are packaged with a variety of signalling molecules, mediating cell-cell communications. Within the TME, exosomes can originate from tumour, immune, or stromal cells. Although there are studies on tumour-derived exosomes (TEX) in NPC and their effects on tumour processes like angiogenesis, metastasis, therapeutic resistance, there is a lack of research on their involvement in immune evasion. In this review, we aim to enhance the comprehension of how NPC TEX contribute to cellular immunosuppression. Furthermore, considering the detectability of TEX in bodily fluids, we will also discuss the potential development of TEX-related biomarkers for liquid biopsy in NPC as this could facilitate early diagnosis and prognostication of the disease.
Collapse
Affiliation(s)
- Paak-Ting Chak
- Department of Clinical Oncology, Centre of Cancer Medicine, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong 999077, China
| | - Ngar-Woon Kam
- Department of Clinical Oncology, Centre of Cancer Medicine, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong 999077, China
- Laboratory for Synthetic Chemistry and Chemical Biology Limited, Hong Kong Science Park, New Territories, Hong Kong 999077, China
| | - Tsz-Ho Choi
- Department of Clinical Oncology, Centre of Cancer Medicine, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong 999077, China
| | - Wei Dai
- Department of Clinical Oncology, Centre of Cancer Medicine, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong 999077, China
- Clinical Oncology Center, The University of Hong Kong-Shenzhen Hospital, Shenzhen 518053, China
| | - Dora Lai-Wan Kwong
- Department of Clinical Oncology, Centre of Cancer Medicine, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong 999077, China
- Clinical Oncology Center, The University of Hong Kong-Shenzhen Hospital, Shenzhen 518053, China
| |
Collapse
|
11
|
Boddu VK, Zamzow P, Kramer MW, Merseburger AS, Gorantla SP, Klinger M, Cramer L, Sauer T, Gemoll T, von Bubnoff N, Gieseler F, Darabi M. Targeting cancer-derived extracellular vesicles by combining CD147 inhibition with tissue factor pathway inhibitor for the management of urothelial cancer cells. Cell Commun Signal 2024; 22:129. [PMID: 38360687 PMCID: PMC10870545 DOI: 10.1186/s12964-024-01508-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 01/31/2024] [Indexed: 02/17/2024] Open
Abstract
BACKGROUND Extracellular vesicles (EVs), including microvesicles, hold promise for the management of bladder urothelial carcinoma (BLCA), particularly because of their utility in identifying therapeutic targets and their diagnostic potential using easily accessible urine samples. Among the transmembrane glycoproteins highly enriched in cancer-derived EVs, tissue factor (TF) and CD147 have been implicated in promoting tumor progression. In this in vitro study, we explored a novel approach to impede cancer cell migration and metastasis by simultaneously targeting these molecules on urothelial cancer-derived EVs. METHODS Cell culture supernatants from invasive and non-invasive bladder cancer cell lines and urine samples from patients with BLCA were collected. Large, microvesicle-like EVs were isolated using sequential centrifugation and characterized by electron microscopy, nanoparticle tracking analysis, and flow cytometry. The impact of urinary or cell supernatant-derived EVs on cellular phenotypes was evaluated using cell-based assays following combined treatment with a specific CD147 inhibitor alone or in combination with a tissue factor pathway inhibitor (TFPI), an endogenous anticoagulant protein that can be released by low-molecular-weight heparins. RESULTS We observed that EVs obtained from the urine samples of patients with muscle-invasive BLCA and from the aggressive bladder cancer cell line J82 exhibited higher TF activity and CD147 expression levels than did their non-invasive counterparts. The shedding of GFP-tagged CD147 into isolated vesicles demonstrated that the vesicles originated from plasma cell membranes. EVs originating from invasive cancer cells were found to trigger migration, secretion of matrix metalloproteinases (MMPs), and invasion. The same induction of MMP activity was replicated using EVs obtained from urine samples of patients with invasive BLCA. EVs derived from cancer cell clones overexpressing TF and CD147 were produced in higher quantities and exhibited a higher invasive potential than those from control cancer cells. TFPI interfered with the effect when used in conjunction with the CD147 inhibitor, further suppressing homotypic EV-induced migration, MMP production, and invasion. CONCLUSIONS Our findings suggest that combining a CD147 inhibitor with low molecular weight heparins to induce TFPI release may be a promising therapeutic approach for urothelial cancer management. This combination can potentially suppress the tumor-promoting actions of cancer-derived microvesicle-like EVs, including collective matrix invasion.
Collapse
Affiliation(s)
- Vijay Kumar Boddu
- Department of Hematology and Oncology, Section for Experimental Oncology, University Hospital Schleswig-Holstein, Lübeck, Germany
| | - Piet Zamzow
- Department of Hematology and Oncology, Section for Experimental Oncology, University Hospital Schleswig-Holstein, Lübeck, Germany
| | | | - Axel S Merseburger
- Department of Urology, University Hospital Schleswig-Holstein, Lübeck, Germany
| | | | | | - Lena Cramer
- Department of Hematology and Oncology, Section for Experimental Oncology, University Hospital Schleswig-Holstein, Lübeck, Germany
| | - Thorben Sauer
- Department of Surgery, Section for Translational Surgical Oncology and Biobanking, University Hospital Schleswig-Holstein, Lübeck, Germany
| | - Timo Gemoll
- Department of Surgery, Section for Translational Surgical Oncology and Biobanking, University Hospital Schleswig-Holstein, Lübeck, Germany
| | - Nikolas von Bubnoff
- Department of Urology, University Hospital Schleswig-Holstein, Lübeck, Germany
- University Cancer Center Schleswig-Holstein (UCCSH), Lübeck, Germany
| | - Frank Gieseler
- Department of Hematology and Oncology, Section for Experimental Oncology, University Hospital Schleswig-Holstein, Lübeck, Germany
- University Cancer Center Schleswig-Holstein (UCCSH), Lübeck, Germany
| | - Masoud Darabi
- Department of Hematology and Oncology, Section for Experimental Oncology, University Hospital Schleswig-Holstein, Lübeck, Germany.
- University Cancer Center Schleswig-Holstein (UCCSH), Lübeck, Germany.
| |
Collapse
|
12
|
Liu Q, Bode AM, Chen X, Luo X. Metabolic reprogramming in nasopharyngeal carcinoma: Mechanisms and therapeutic opportunities. Biochim Biophys Acta Rev Cancer 2023; 1878:189023. [PMID: 37979733 DOI: 10.1016/j.bbcan.2023.189023] [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: 09/25/2023] [Revised: 11/08/2023] [Accepted: 11/09/2023] [Indexed: 11/20/2023]
Abstract
The high prevalence of metabolic reprogramming in nasopharyngeal carcinoma (NPC) offers an abundance of potential therapeutic targets. This review delves into the distinct mechanisms underlying metabolic reprogramming in NPC, including enhanced glycolysis, nucleotide synthesis, and lipid metabolism. All of these changes are modulated by Epstein-Barr virus (EBV) infection, hypoxia, and tumor microenvironment. We highlight the role of metabolic reprogramming in the development of NPC resistance to standard therapies, which represents a challenging barrier in treating this malignancy. Furthermore, we dissect the state of the art in therapeutic strategies that target these metabolic changes, evaluating the successes and failures of clinical trials and the strategies to tackle resistance mechanisms. By providing a comprehensive overview of the current knowledge and future directions in this field, this review sets the stage for new therapeutic avenues in NPC.
Collapse
Affiliation(s)
- Qian Liu
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Oncotarget Gene, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, PR China; Key Laboratory of Carcinogenesis and Invasion, Chinese Ministry of Education, Cancer Research Institute, School of Basic Medicine, Central South University, Changsha, Hunan 410078, PR China
| | - Ann M Bode
- The Hormel Institute, University of Minnesota, Austin, MN 55912, USA
| | - Xue Chen
- Early Clinical Trial Center, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, PR China.
| | - Xiangjian Luo
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Oncotarget Gene, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, PR China; Key Laboratory of Carcinogenesis and Invasion, Chinese Ministry of Education, Cancer Research Institute, School of Basic Medicine, Central South University, Changsha, Hunan 410078, PR China; Key Laboratory of Biological Nanotechnology of National Health Commission, Central South University, Changsha, Hunan 410078, China.
| |
Collapse
|
13
|
Wang X, Zhang Y, Chung Y, Tu CR, Zhang W, Mu X, Wang M, Chan GC, Leung W, Lau Y, Liu Y, Tu W. Tumor vaccine based on extracellular vesicles derived from γδ-T cells exerts dual antitumor activities. J Extracell Vesicles 2023; 12:e12360. [PMID: 37654012 PMCID: PMC10471836 DOI: 10.1002/jev2.12360] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Accepted: 08/09/2023] [Indexed: 09/02/2023] Open
Abstract
γδ-T cells are innate-like T cells with dual antitumor activities. They can directly eradicate tumor cells and function as immunostimulatory cells to promote antitumor immunity. Previous studies have demonstrated that small extracellular vesicles (EVs) derived from γδ-T cells (γδ-T-EVs) inherited the dual antitumor activities from their parental cells. However, it remains unknown whether γδ-T-EVs can be designed as tumors vaccine to improve therapeutic efficacy. Here, we found that γδ-T-EVs had immune adjuvant effects on antigen-presenting cells, as revealed by enhanced expression of antigen-presenting and co-stimulatory molecules, secretion of pro-inflammatory cytokines and antigen-presenting ability of DCs after γδ-T-EVs treatment. The γδ-T-EVs-based vaccine was designed by loading tumor-associated antigens (TAAs) into γδ-T-EVs. Compared with γδ-T-EVs, the γδ-T-EVs-based vaccine effectively promoted more tumor-specific T-cell responses. In addition, the vaccine regimen preserved direct antitumor effects and induced tumor cell apoptosis. Interestingly, the allogeneic γδ-T-EVs-based vaccine showed comparable preventive and therapeutic antitumor effects to their autologous counterparts, indicating a better way of centralization and standardization in clinical practice. Furthermore, the allogeneic γδ-T-EVs-based vaccine displayed advantages over the DC-EVs-based vaccine through their dual antitumor activities. This study provides a proof-of-concept for using the allogeneic γδ-T-EVs-based vaccine in cancer control.
Collapse
Affiliation(s)
- Xiwei Wang
- Department of Paediatrics & Adolescent Medicine, Li Ka Shing Faculty of MedicineThe University of Hong KongHong Kong SARChina
| | - Yanmei Zhang
- Department of Paediatrics & Adolescent Medicine, Li Ka Shing Faculty of MedicineThe University of Hong KongHong Kong SARChina
| | - Yuet Chung
- Department of Paediatrics & Adolescent Medicine, Li Ka Shing Faculty of MedicineThe University of Hong KongHong Kong SARChina
| | - Chloe Ran Tu
- Department of Data Sciences, Dana‐Farber Cancer InstituteHarvard UniversityBostonMassachusettsUSA
| | - Wenyue Zhang
- Department of Paediatrics & Adolescent Medicine, Li Ka Shing Faculty of MedicineThe University of Hong KongHong Kong SARChina
| | - Xiaofeng Mu
- Department of Paediatrics & Adolescent Medicine, Li Ka Shing Faculty of MedicineThe University of Hong KongHong Kong SARChina
| | - Manni Wang
- Department of Paediatrics & Adolescent Medicine, Li Ka Shing Faculty of MedicineThe University of Hong KongHong Kong SARChina
| | - Godfrey Chi‐Fung Chan
- Department of Paediatrics & Adolescent Medicine, Li Ka Shing Faculty of MedicineThe University of Hong KongHong Kong SARChina
| | - Wing‐Hang Leung
- Department of Paediatrics & Adolescent Medicine, Li Ka Shing Faculty of MedicineThe University of Hong KongHong Kong SARChina
| | - Yu‐Lung Lau
- Department of Paediatrics & Adolescent Medicine, Li Ka Shing Faculty of MedicineThe University of Hong KongHong Kong SARChina
| | - Yinping Liu
- Department of Paediatrics & Adolescent Medicine, Li Ka Shing Faculty of MedicineThe University of Hong KongHong Kong SARChina
| | - Wenwei Tu
- Department of Paediatrics & Adolescent Medicine, Li Ka Shing Faculty of MedicineThe University of Hong KongHong Kong SARChina
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced TechnologyChinese Academy of SciencesShenzhenChina
| |
Collapse
|
14
|
Zhang J, Huang D, Lan X, Deng D, Li J, Zhang D, Li Y, Zhong T, Peng S. Application of small extracellular vesicles in the diagnosis and prognosis of nasopharyngeal carcinoma. Front Cell Dev Biol 2023; 11:1100941. [PMID: 36968209 PMCID: PMC10036369 DOI: 10.3389/fcell.2023.1100941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 03/02/2023] [Indexed: 03/12/2023] Open
Abstract
Nasopharyngeal carcinoma (NPC) is a malignant tumor originating from the epithelium of the nasopharynx. The disease is insidious, and most patients are diagnosed at the advanced stage, resulting in poor prognosis. Early diagnosis is important to reduce NPC mortality. Small extracellular vesicles (sEVs) are rich in a variety of bioactive molecules, such as proteins, nucleic acids, and lipids, which can participate in the physiological and pathological regulation of the body by affecting the function of target cells. Numerous studies have shown that some RNAs and proteins in sEVs of tumor origin have a key role in the development of NPC and are potential candidates for malignancy detection. Studying the relationship between the cargoes of these sEVs and NPC may help in the diagnosis of the disease. Here in this review, we summarize the application of sEVs as biomarkers in the diagnosis of NPC and their role in NPC metastasis and prognosis. In addition, we discuss possible future applications and limitations of sEVs as biomarkers.
Collapse
Affiliation(s)
- Jiali Zhang
- The First School of Clinical Medicine, Gannan Medical University, Ganzhou, China
- Department of Otolaryngology, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Defa Huang
- The First School of Clinical Medicine, Gannan Medical University, Ganzhou, China
- Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Xianbin Lan
- The First School of Clinical Medicine, Gannan Medical University, Ganzhou, China
- Department of Otolaryngology, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Dongming Deng
- The First School of Clinical Medicine, Gannan Medical University, Ganzhou, China
- Department of Otolaryngology, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Jijing Li
- The First School of Clinical Medicine, Gannan Medical University, Ganzhou, China
- Department of Otolaryngology, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Dongzhi Zhang
- The First School of Clinical Medicine, Gannan Medical University, Ganzhou, China
- Department of Otolaryngology, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Yue Li
- The First School of Clinical Medicine, Gannan Medical University, Ganzhou, China
- Department of Otolaryngology, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Tianyu Zhong
- The First School of Clinical Medicine, Gannan Medical University, Ganzhou, China
- Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
- Precision Medicine Center, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
- *Correspondence: Tianyu Zhong, ; Shaoping Peng,
| | - Shaoping Peng
- The First School of Clinical Medicine, Gannan Medical University, Ganzhou, China
- Department of Otolaryngology, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
- *Correspondence: Tianyu Zhong, ; Shaoping Peng,
| |
Collapse
|
15
|
Ding J, Xiao R, Bi A, Chen G, Zhang N, Chen Z, Feng X, Zeng W. An ESIPT-based NIR-fluorescent probe for exosome labelling and in situ imaging. CHINESE CHEM LETT 2023. [DOI: 10.1016/j.cclet.2023.108273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2023]
|
16
|
Yeung CLS, Yam JWP. Therapy-induced modulation of extracellular vesicles in hepatocellular carcinoma. Semin Cancer Biol 2022; 86:1088-1101. [PMID: 35158067 DOI: 10.1016/j.semcancer.2022.02.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 02/09/2022] [Accepted: 02/09/2022] [Indexed: 01/27/2023]
Abstract
Despite rapid development of anti-tumorigenic treatments, the clinical outcome for hepatocellular carcinoma (HCC) is still far from satisfactory. With a deeper understanding about tumor microenvironment (TME), the critical role of extracellular vesicles (EVs) as intercellular liaison has come into spotlight. The dynamic functionality of these nanoparticles revealed cancer cells can employ both tumor and non-tumorous components for their own benefit, so as to mediate cell-to-cell communication and interchange of oncogenic biomolecules. Increasing studies on HCC-derived EVs have identified various irregulated biomolecules, that may serve as biomarkers or therapeutic targets. In this review, we first introduce the current knowledge about EVs and how they operate to maintain a healthy liver microenvironment. We then summarize some of the aberrant observations reported on HCC-derived EVs and how they contribute to HCC pathogenesis. Finally, we describe how current treatments for HCC alter behavior of EVs, which may shed light for potential prognostic markers and therapeutic strategies.
Collapse
Affiliation(s)
- Cherlie Lot Sum Yeung
- Department of Pathology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong
| | - Judy Wai Ping Yam
- Department of Pathology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong; State Key Laboratory of Liver Research (The University of Hong Kong), Hong Kong.
| |
Collapse
|
17
|
Song F, Wang C, Wang C, Wang J, Wu Y, Wang Y, Liu H, Zhang Y, Han L. Multi-Phenotypic Exosome Secretion Profiling Microfluidic Platform for Exploring Single-Cell Heterogeneity. SMALL METHODS 2022; 6:e2200717. [PMID: 35901289 DOI: 10.1002/smtd.202200717] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Indexed: 06/15/2023]
Abstract
Cellular phenotypic and functional heterogeneities have advanced cancer evolution and treatment resistance. Although exosome-bound proteins reflect cellular functions, single-cell exosomes are rarely profiled owing to the lack of effective platforms. Herein, the authors developed an integrated microfluidic platform consisting of a single-cell trapping chip and a spatially coded antibody barcode chip for the multiplexed outline of exosome secretion by single cells. Using this platform, five phenotypic exosomes of over 1 000 single cells are simultaneously profiled, in addition to inflammatory factor secretion from the same single cell. Also, a robust analysis workflow for single-cell secretion profiling is proposed to explore the intercellular heterogeneity, which integrated unsupervised clustering and linear clustering. When applied to the tumor cell lines of epithelial-origin and normal epithelial cell lines, the strategy identifies functionally heterogeneous subpopulations with unique secretion patterns. Notably, special functional cell subsets for unique phenotypic exosomes (HSP70+ , EPCAM+ ) are found within ovarian tumor cells. The strategy proposed offers a new analysis approach for cellular differential exosome secretion at single-cell resolution using inflammatory factors, ultimately reinforcing the understanding of cell-to-cell heterogeneity and tumor landscape, and providing a valuable universal platform for single-cell biomarker exploration in biological and clinical research.
Collapse
Affiliation(s)
- Fangteng Song
- Institute of Marine Science and Technology, Shandong University, Qingdao, Shandong, 266000, China
| | - Chao Wang
- Institute of Marine Science and Technology, Shandong University, Qingdao, Shandong, 266000, China
| | - Chunhua Wang
- Institute of Marine Science and Technology, Shandong University, Qingdao, Shandong, 266000, China
| | - Jianbo Wang
- Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250100, China
| | - Yu Wu
- Peking University Third Hospital, Peking University, Beijing, 100191, China
| | - Yihe Wang
- Institute of Marine Science and Technology, Shandong University, Qingdao, Shandong, 266000, China
| | - Hong Liu
- State Key Laboratory of Crystal Materials, Center of Bio & Micro/Nano Functional Materials, Shandong University, Jinan, Shandong, 250100, China
| | - Yu Zhang
- Institute of Marine Science and Technology, Shandong University, Qingdao, Shandong, 266000, China
- State Key Laboratory of Microbial Technology, Qingdao, Shandong, 266000, China
| | - Lin Han
- Institute of Marine Science and Technology, Shandong University, Qingdao, Shandong, 266000, China
- Shandong Engineering Research Center of Biomarker and Artificial Intelligence Application, Jinan, Shandong, 250100, China
| |
Collapse
|
18
|
Jiang J, Ying H. Revealing the crosstalk between nasopharyngeal carcinoma and immune cells in the tumor microenvironment. J Exp Clin Cancer Res 2022; 41:244. [PMID: 35964134 PMCID: PMC9375932 DOI: 10.1186/s13046-022-02457-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 08/01/2022] [Indexed: 01/13/2023] Open
Abstract
Nasopharyngeal carcinoma (NPC) arises from the epithelial cells located in the nasopharynx and has a distinct geographic distribution. Chronic Epstein-Barr virus (EBV) infection, as its most common causative agents, can be detected in 100% of NPC types. In-depth studies of the cellular and molecular events leading to immunosuppression in NPC have revealed new therapeutic targets and diverse combinations that promise to benefit patients with highly refractory, advanced and metastatic NPC. This paper reviews the mechanisms by which NPC cells to circumvent immune surveillance and approaches being attempted to restore immunity. We integrate existing insights into anti-NPC immunity and molecular signaling pathways as well as targeting therapies in anticipation of broader applicability and effectiveness in advanced metastatic NPC.
Collapse
|
19
|
Shan Y, Zhou P, Zhou Q, Yang L. Extracellular Vesicles in the Progression and Therapeutic Resistance of Nasopharyngeal Carcinoma. Cancers (Basel) 2022; 14:2289. [PMID: 35565418 PMCID: PMC9101631 DOI: 10.3390/cancers14092289] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Revised: 04/30/2022] [Accepted: 05/02/2022] [Indexed: 02/07/2023] Open
Abstract
Nasopharyngeal carcinoma (NPC) is an epithelial malignancy largely associated with Epstein-Barr virus (EBV) infection, which is frequently reported in east and southeast Asia. Extracellular vesicles (EVs) originate from the endosome or plasma membrane, which plays a critical role in tumor pathogenesis for their character of cell-cell communication and its cargos, including proteins, RNA, and other molecules that can target recipient cells and affect their progression. To date, numerous studies have indicated that EVs have crucial significance in the progression, metastasis, and therapeutic resistance of NPC. In this review, we not only summarize the interaction of NPC cells and the tumor microenvironment (TME) through EVs, but also explain the role of EVs in radiation and drug resistance of NPC, which poses a severe threat to cancer therapy. Therefore, EVs may show great potential as biomarkers in the early diagnosis of interfered targets of NPC therapy.
Collapse
Affiliation(s)
- Yunhan Shan
- Department of Oncology, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410078, China; (Y.S.); (P.Z.); (Q.Z.)
- Cancer Research Institute, School of Basic Medicine Science, Central South University, Changsha 410078, China
- Xiangya School of Medicine, Central South University, Changsha 410013, China
| | - Peijun Zhou
- Department of Oncology, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410078, China; (Y.S.); (P.Z.); (Q.Z.)
- Cancer Research Institute, School of Basic Medicine Science, Central South University, Changsha 410078, China
| | - Qin Zhou
- Department of Oncology, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410078, China; (Y.S.); (P.Z.); (Q.Z.)
| | - Lifang Yang
- Department of Oncology, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410078, China; (Y.S.); (P.Z.); (Q.Z.)
- Cancer Research Institute, School of Basic Medicine Science, Central South University, Changsha 410078, China
| |
Collapse
|
20
|
Su ZY, Siak PY, Leong CO, Cheah SC. Nasopharyngeal Carcinoma and Its Microenvironment: Past, Current, and Future Perspectives. Front Oncol 2022; 12:840467. [PMID: 35311066 PMCID: PMC8924466 DOI: 10.3389/fonc.2022.840467] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 02/11/2022] [Indexed: 12/31/2022] Open
Abstract
Nasopharyngeal carcinoma (NPC) is an epithelial malignancy that raises public health concerns in endemic countries. Despite breakthroughs in therapeutic strategies, late diagnosis and drug resistance often lead to unsatisfactory clinical outcomes in NPC patients. The tumor microenvironment (TME) is a complex niche consisting of tumor-associated cells, such as fibroblasts, endothelial cells, leukocytes, that influences tumor initiation, progression, invasion, and metastasis. Cells in the TME communicate through various mechanisms, of note, exosomes, ligand-receptor interactions, cytokines and chemokines are active players in the construction of TME, characterized by an abundance of immune infiltrates with suppressed immune activities. The NPC microenvironment serves as a target-rich niche for the discovery of potential promising predictive or diagnostic biomarkers and the development of therapeutic strategies. Thus, huge efforts have been made to exploit the role of the NPC microenvironment. The whole picture of the NPC microenvironment remains to be portrayed to understand the mechanisms underlying tumor biology and implement research into clinical practice. The current review discusses the recent insights into the role of TME in the development and progression of NPC which results in different clinical outcomes of patients. Clinical interventions with the use of TME components as potential biomarkers or therapeutic targets, their challenges, and future perspectives will be introduced. This review anticipates to provide insights to the researchers for future preclinical, translational and clinical research on the NPC microenvironment.
Collapse
Affiliation(s)
- Zhi Yi Su
- Faculty of Medicine and Health Sciences, UCSI University, Kuala Lumpur, Malaysia
| | - Pui Yan Siak
- Faculty of Medicine and Health Sciences, UCSI University, Kuala Lumpur, Malaysia
| | - Chee-Onn Leong
- Centre of Cancer and Stem Cells Research, International Medical University, Kuala Lumpur, Malaysia
- Institute for Research, Development and Innovation, International Medical University, Kuala Lumpur, Malaysia
| | - Shiau-Chuen Cheah
- Faculty of Medicine and Health Sciences, UCSI University, Kuala Lumpur, Malaysia
| |
Collapse
|
21
|
Wang X, Zhang Y, Mu X, Tu CR, Chung Y, Tsao SW, Chan GCF, Leung WH, Lau YL, Liu Y, Tu W. Exosomes derived from γδ-T cells synergize with radiotherapy and preserve antitumor activities against nasopharyngeal carcinoma in immunosuppressive microenvironment. J Immunother Cancer 2022; 10:jitc-2021-003832. [PMID: 35105688 PMCID: PMC8808451 DOI: 10.1136/jitc-2021-003832] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/04/2022] [Indexed: 12/30/2022] Open
Abstract
Background Radiotherapy is the first-line treatment for patients nasopharyngeal carcinoma (NPC), but its therapeutic efficacy is poor in some patients due to radioresistance. Adoptive T cell-based immunotherapy has also shown promise to control NPC; however, its antitumor efficacy may be attenuated by an immunosuppressive tumor microenvironment. Exosomes derived from γδ-T cells (γδ-T-Exos) have potent antitumor potentials. However, it remains unknown whether γδ-T-Exos have synergistic effect with radiotherapy and preserve their antitumor activities against NPC in an immunosuppressive tumor microenvironment. Methods γδ-T-Exos were stained with fluorescent membrane dye, and their interactions with NPC were determined both in vitro and in vivo. NPC cell deaths were detected after treatment with γδ-T-Exos and/or irradiation. Moreover, effects of γδ-T-Exos on radioresistant cancer stem-like cells (CSCs) were determined. The therapeutic efficacy of combination therapy using γδ-T-Exos and irradiation on NPC tumor progression was also monitored in vivo. Finally, the tumor-killing and T cell-promoting activities of γδ-T-Exos were determined under the culture in immunosuppressive NPC supernatant. Results γδ-T-Exos effectively interacted with NPC tumor cells in vitro and in vivo. γδ-T-Exos not only killed NPC cells in vitro, which was mainly mediated by Fas/Fas ligand (FasL) and death receptor 5 (DR5)/tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) pathways, but also controlled NPC tumor growth and prolonged tumor-bearing mice survival in vivo. Furthermore, γδ-T-Exos selectively targeted the radioresistant CD44+/high CSCs and induced profound cell apoptosis. The combination of γδ-T-Exos with radiotherapy overcame the radioresistance of CD44+/high NPC cells and significantly improved its therapeutic efficacy against NPC in vitro and in vivo. In addition, γδ-T-Exos promoted T-cell migration into NPC tumors by upregulating CCR5 on T cells that were chemoattracted by CCR5 ligands in the NPC tumor microenvironment. Although NPC tumor cells secreted abundant tumor growth factor beta to suppress T-cell responses, γδ-T-Exos preserved their direct antitumor activities and overcame the immunosuppressive NPC microenvironment to amplify T-cell antitumor immunity. Conclusions γδ-T-Exos synergized with radiotherapy to control NPC by overcoming the radioresistance of NPC CSCs. Moreover, γδ-T-Exos preserved their tumor-killing and T cell-promoting activities in the immunosuppressive NPC microenvironment. This study provides a proof of concept for a novel and potent strategy by combining γδ-T-Exos with radiotherapy in the control of NPC.
Collapse
Affiliation(s)
- Xiwei Wang
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Yanmei Zhang
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Xiaofeng Mu
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Chloe Ran Tu
- Computational and Systems Biology Interdepartmental Program, University of California Los Angeles, Los Angeles, California, USA
| | - Yuet Chung
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Sai Wah Tsao
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong, China
| | - Godfrey Chi-Fung Chan
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Wing-Hang Leung
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Yu-Lung Lau
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Yinping Liu
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Wenwei Tu
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| |
Collapse
|
22
|
Chen P, Liu R, Yu Z, Cui G, Zong W, Wang M, Xie M, Qu W, Wang W, Luo X. MiR196a-5p in extracellular vesicles released from human nasopharyngeal carcinoma enhance the phagocytosis and secretion of microglia by targeting ROCK1. Exp Cell Res 2021; 411:112988. [PMID: 34951996 DOI: 10.1016/j.yexcr.2021.112988] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 12/15/2021] [Accepted: 12/19/2021] [Indexed: 12/19/2022]
Abstract
The microenvironment of the brain has become increasingly recognized as an essential regulator in metastatic and primary brain tumors. Recent studies demonstrate that circulating tumor-derived exosomes are critical for the brain tumor microenvironment. Nasopharyngeal carcinoma (NPC), a malignant tumor of the head and neck, often invades the skull base but infrequently extends to brain parenchyma. Neurobiological communication between microglia and tumor-derived extracellular vesicles (EVs) has been extensively studied, but how NPC cells regulate the immune microenvironment in the brain remains unknown. Here, we report that NPC derived EVs lead to increased microglial phagocytosis and proliferation, and heightened levels of IL-6, IL-8, CXCL1 and TGF-β1. Analysis of microRNAs in EVs reveal that miR196a-5p is the major effector microRNA. Moreover, we demonstrate an enrichment of miR196a-5p in the plasmatic EVs of NPC patients. Further investigation demonstrated that miR196a-5p was transferred to microglia and regulated microglial structure and functions by downregulating the expression of ROCK1. Therefore, these data indicate that NPC-derived EVs are potent modulators of microglial functions in brain microenvironment. Regardless of brain colonization, EVs-mediated functional changes in microglia may be a universal phenomenon that results in the alteration of the tumor host's microenvironment in the brain.
Collapse
Affiliation(s)
- Peng Chen
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Liberation Avenue, Wuhan, 430030, China
| | - Rui Liu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Liberation Avenue, Wuhan, 430030, China
| | - Zhiyuan Yu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Liberation Avenue, Wuhan, 430030, China
| | - GuoHui Cui
- Department of Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, No. 107 Yan Jiang West Road, Guangzhou, 510120, China
| | - Weifeng Zong
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Liberation Avenue, Wuhan, 430030, China
| | - Minghuan Wang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Liberation Avenue, Wuhan, 430030, China
| | - Minjie Xie
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Liberation Avenue, Wuhan, 430030, China
| | - Wensheng Qu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Liberation Avenue, Wuhan, 430030, China
| | - Wei Wang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Liberation Avenue, Wuhan, 430030, China
| | - Xiang Luo
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Liberation Avenue, Wuhan, 430030, China.
| |
Collapse
|
23
|
Luo H, Yi B. The role of Exosomes in the Pathogenesis of Nasopharyngeal Carcinoma and the involved Clinical Application. Int J Biol Sci 2021; 17:2147-2156. [PMID: 34239345 PMCID: PMC8241729 DOI: 10.7150/ijbs.59688] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 05/17/2021] [Indexed: 12/12/2022] Open
Abstract
Exosomes are nanoscale membrane vesicles, which carry biologically active substances of their cell of origin and play an important role in signal transduction and intercellular communication. At present, exosomes have been identified as a promising non-invasive liquid biopsy biomarker in the tissues and circulating blood of nasopharyngeal carcinoma (NPC) and found to participate in regulating pathophysiological process of the tumor. We here review recent insights gained into the molecular mechanisms of exosome-induced cell growth, angiogenesis, metastasis, immunosuppression, radiation resistance and chemotherapy resistance in the development and progression of NPC, as well as the clinical application of exosomes as diagnostic biomarkers and therapeutic agents. We also discuss the limitations and challenges in exosome application. We hope this review may provide some references for the use of exosomes in clinical intervention.
Collapse
Affiliation(s)
- Huidan Luo
- Department of Clinical Laboratory, Xiangya Hospital, Central South University, Changsha, Hunan Province 410008, China
| | - Bin Yi
- Department of Clinical Laboratory, Xiangya Hospital, Central South University, Changsha, Hunan Province 410008, China
| |
Collapse
|
24
|
Bai K, Li X, Zhong J, Ng EHY, Yeung WSB, Lee CL, Chiu PCN. Placenta-Derived Exosomes as a Modulator in Maternal Immune Tolerance During Pregnancy. Front Immunol 2021; 12:671093. [PMID: 34046039 PMCID: PMC8144714 DOI: 10.3389/fimmu.2021.671093] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 04/26/2021] [Indexed: 12/12/2022] Open
Abstract
Exosomes are a subset of extracellular vesicles with an average diameter of ~100nm. Exosomes are released by all cells through an endosome-dependent pathway and carry nucleic acids, proteins, lipids, cytokines and metabolites, mirroring the state of the originating cells. The function of exosomes has been implicated in various reproduction processes, such as embryo development, implantation, decidualization and placentation. Placenta-derived exosomes (pEXO) can be detected in the maternal blood as early as 6 weeks after conception and their levels increase with gestational age. Importantly, alternations in the molecular signatures of pEXO are observed in pregnancy-related complications. Thus, these differentially expressed molecules could be the potential biomarkers for diagnosis of the pregnancy-associated diseases. Recent studies have demonstrated that pEXO play a key role in the establishment of maternal immune tolerance, which is critical for a successful pregnancy. To gain a better understanding of the underlying mechanism, we highlighted the advanced studies of pEXO on immune cells in pregnancy.
Collapse
Affiliation(s)
- Kunfeng Bai
- Department of Obstetrics and Gynaecology, Li Ka Shing (LKS) Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong.,The University of Hong Kong Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Xintong Li
- Department of Obstetrics and Gynaecology, Li Ka Shing (LKS) Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong
| | - Jiangming Zhong
- Department of Obstetrics and Gynaecology, Li Ka Shing (LKS) Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong
| | - Ernest H Y Ng
- Department of Obstetrics and Gynaecology, Li Ka Shing (LKS) Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong.,The University of Hong Kong Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - William S B Yeung
- The University of Hong Kong Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Cheuk-Lun Lee
- Department of Obstetrics and Gynaecology, Li Ka Shing (LKS) Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong.,The University of Hong Kong Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Philip C N Chiu
- Department of Obstetrics and Gynaecology, Li Ka Shing (LKS) Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong.,The University of Hong Kong Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| |
Collapse
|
25
|
Longo V, Longo A, Adamo G, Fiannaca A, Picciotto S, La Paglia L, Romancino D, La Rosa M, Urso A, Cibella F, Bongiovanni A, Colombo P. 2,2'4,4'-Tetrabromodiphenyl Ether (PBDE-47) Modulates the Intracellular miRNA Profile, sEV Biogenesis and Their miRNA Cargo Exacerbating the LPS-Induced Pro-Inflammatory Response in THP-1 Macrophages. Front Immunol 2021; 12:664534. [PMID: 34025666 PMCID: PMC8138315 DOI: 10.3389/fimmu.2021.664534] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 04/22/2021] [Indexed: 12/30/2022] Open
Abstract
The 2,2’4,4’-tetrabromodiphenyl ether (PBDE-47) is one of the most prominent PBDE congeners detected in the environment and in animal and human tissues. Animal model experiments suggested the occurrence of PBDE-induced immunotoxicity leading to different outcomes and recently we demonstrated that this substance can impair macrophage and basophil activities. In this manuscript, we decided to further examine the effects induced by PBDE-47 treatment on innate immune response by looking at the intracellular expression profile of miRNAs as well as the biogenesis, cargo content and activity of human M(LPS) macrophage cell-derived small extracellular vesicles (sEVs). Microarray and in silico analysis demonstrated that PBDE-47 can induce some epigenetic effects in M(LPS) THP-1 cells modulating the expression of a set of intracellular miRNAs involved in biological pathways regulating the expression of estrogen-mediated signaling and immune responses with particular reference to M1/M2 differentiation. In addition to the cell-intrinsic modulation of intracellular miRNAs, we demonstrated that PBDE-47 could also interfere with the biogenesis of sEVs increasing their number and selecting a de novo population of sEVs. Moreover, PBDE-47 induced the overload of specific immune related miRNAs in PBDE-47 derived sEVs. Finally, culture experiments with naïve M(LPS) macrophages demonstrated that purified PBDE-47 derived sEVs can modulate macrophage immune response exacerbating the LPS-induced pro-inflammatory response inducing the overexpression of the IL-6 and the MMP9 genes. Data from this study demonstrated that PBDE-47 can perturb the innate immune response at different levels modulating the intracellular expression of miRNAs but also interfering with the biogenesis, cargo content and functional activity of M(LPS) macrophage cell-derived sEVs.
Collapse
Affiliation(s)
- Valeria Longo
- Institute for Biomedical Research and Innovation, National Research Council of Italy (IRIB-CNR), Palermo, Italy
| | - Alessandra Longo
- Institute for Biomedical Research and Innovation, National Research Council of Italy (IRIB-CNR), Palermo, Italy
| | - Giorgia Adamo
- Institute for Biomedical Research and Innovation, National Research Council of Italy (IRIB-CNR), Palermo, Italy
| | - Antonino Fiannaca
- High Performance Computing and Networking Institute, National Research Council of Italy (ICAR-CNR), Palermo, Italy
| | - Sabrina Picciotto
- Institute for Biomedical Research and Innovation, National Research Council of Italy (IRIB-CNR), Palermo, Italy
| | - Laura La Paglia
- High Performance Computing and Networking Institute, National Research Council of Italy (ICAR-CNR), Palermo, Italy
| | - Daniele Romancino
- Institute for Biomedical Research and Innovation, National Research Council of Italy (IRIB-CNR), Palermo, Italy
| | - Massimo La Rosa
- High Performance Computing and Networking Institute, National Research Council of Italy (ICAR-CNR), Palermo, Italy
| | - Alfonso Urso
- High Performance Computing and Networking Institute, National Research Council of Italy (ICAR-CNR), Palermo, Italy
| | - Fabio Cibella
- Institute for Biomedical Research and Innovation, National Research Council of Italy (IRIB-CNR), Palermo, Italy
| | - Antonella Bongiovanni
- Institute for Biomedical Research and Innovation, National Research Council of Italy (IRIB-CNR), Palermo, Italy
| | - Paolo Colombo
- Institute for Biomedical Research and Innovation, National Research Council of Italy (IRIB-CNR), Palermo, Italy
| |
Collapse
|
26
|
Zhong Y, Wang D, Ding Y, Tian G, Jiang B. Circular RNA circ_0032821 contributes to oxaliplatin (OXA) resistance of gastric cancer cells by regulating SOX9 via miR-515-5p. Biotechnol Lett 2020; 43:339-351. [PMID: 33123829 DOI: 10.1007/s10529-020-03036-3] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 10/22/2020] [Indexed: 12/24/2022]
Abstract
OBJECTIVES Chemoresistance is one of the major obstacles for gastric cancer (GC) treatment. Exosome-mediated transfer of circular RNAs (circRNAs) is associated with the drug-resistance in GC. Circ_0032821 has been reported as an oncogene in GC. This study is designed to explore the function and mechanism of Exosomal circ_0032821 in oxaliplatin (OXA) resistance of GC. RESULTS Circ_0032821 was highly expressed in OXA-resistant GC cells, and exosomes secreted by OXA-resistant GC cells. Moreover, circ_0032821-containing exosomes secreted by OXA-resistant GC cells could boost OXA resistance, proliferation, migration, and invasion in OXA-sensitive GC cells. The mechanical analysis discovered that circ_0032821 acted as a sponge of miR-515-5p to regulate SOX9 expression. Circ_0032821 silencing and OXA treatment repressed tumor growth in the GC mice model. CONCLUSIONS Exosomal circ_0032821 boosted OXA resistance of GC cells partly by the miR-515-5p/SOX9 axis, hinting a promising therapeutic target for GC treatment.
Collapse
Affiliation(s)
- Yanmei Zhong
- Department of Gastroenterology, Weifang People's Hospital, No. 151, Guangwen Street, Kuiwen District, Weifang, 261041, Shandong, China
| | - Dan Wang
- Department of Gastroenterology, Weifang People's Hospital, No. 151, Guangwen Street, Kuiwen District, Weifang, 261041, Shandong, China
| | - Yanle Ding
- Department of Gastroenterology, Weifang People's Hospital, No. 151, Guangwen Street, Kuiwen District, Weifang, 261041, Shandong, China
| | - Guixin Tian
- Department of Internal Medicine, Changle County Tangwu Town Hospital, Weifang, Shandong, China
| | - Bing Jiang
- Department of Gastroenterology, Weifang People's Hospital, No. 151, Guangwen Street, Kuiwen District, Weifang, 261041, Shandong, China.
| |
Collapse
|