1
|
Li F, Jiang Y, Wang X, Gao Y, Lo CS, Su S, Wu Z, Jiang B, Zhao Z, Lin S, Xie Y, Chen J, Guo Q, Dong Z, Zhu J. Titanium nitride meta-biosensors targeting extracellular vesicles for high-sensitivity prostate cancer detection. Biosens Bioelectron 2025; 277:117288. [PMID: 39985905 DOI: 10.1016/j.bios.2025.117288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 02/16/2025] [Accepted: 02/18/2025] [Indexed: 02/24/2025]
Abstract
Disposable plasmonic metasurfaces with high biosensing performance are urgently sought for clinical label-free detection. Low-cost aluminum (Al) and titanium nitride (TiN) offer promising alternatives to noble metals for constructing these metasurfaces. However, Al suffers from limited chemical stability, and TiN exhibits weak plasmonic effects, both of which hinder their application in meta-biosensing. Here we integrate their complementary advantages and propose the TiN/Al meta-biosensors. They not only empower the unique near-field enhancement for sensing by TiN/Al hybrid plasmonic modes, but also construct a robust TiN armor against external wear, heat, moisture and corrosion during the bio-detection process. Compared to traditional gold-based counterparts, our meta-biosensors offer superior optical sensitivity at a much lower cost and with fewer pretreatment steps. The excellent biosensing performance facilitates the development of a high-throughput detection system for serum small extracellular vesicles (sEVs), aiding in the diagnosis and follow-up of prostate cancer. The sEVs meta-biosensing demonstrates a diagnostic sensitivity of 100% for significantly distinguishing early cancer, breaking through the conventional testing limitation. Moreover, it doubles the prediction accuracy of cancer recurrence risk following surgery. Our research highlights the potential for large-scale development of powerful meta-biosensors based on non-noble materials, opening up significant opportunities in cancer diagnosis and prognosis.
Collapse
Affiliation(s)
- Fajun Li
- Institute of Electromagnetics and Acoustics and Key Laboratory of Electromagnetic Wave Science and Detection Technology, Xiamen University, Xiamen, 361100, China; Xiamen Dili-Chip Technology Co. Ltd., Xiamen, 361000, China; School of Electronic Science and Engineering, National Model Microelectronics College, Xiamen University, Xiamen, 361100, China
| | - Yuanyuan Jiang
- Institute of Electromagnetics and Acoustics and Key Laboratory of Electromagnetic Wave Science and Detection Technology, Xiamen University, Xiamen, 361100, China
| | - Xuegang Wang
- Department of Urology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361003, China
| | - Yuan Gao
- Institute of Electromagnetics and Acoustics and Key Laboratory of Electromagnetic Wave Science and Detection Technology, Xiamen University, Xiamen, 361100, China
| | - Ching Shu Lo
- School of Electronic Science and Engineering, National Model Microelectronics College, Xiamen University, Xiamen, 361100, China
| | - Shengdong Su
- Institute of Electromagnetics and Acoustics and Key Laboratory of Electromagnetic Wave Science and Detection Technology, Xiamen University, Xiamen, 361100, China
| | - Zhilin Wu
- Institute of Electromagnetics and Acoustics and Key Laboratory of Electromagnetic Wave Science and Detection Technology, Xiamen University, Xiamen, 361100, China
| | - Bingliang Jiang
- Department of Urology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361003, China
| | - Zhongjie Zhao
- Department of Urology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361003, China
| | - Shaowei Lin
- Department of Nuclear Medicine, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361003, China
| | - Yinong Xie
- Institute of Electromagnetics and Acoustics and Key Laboratory of Electromagnetic Wave Science and Detection Technology, Xiamen University, Xiamen, 361100, China
| | - Junjie Chen
- Analysis and Measurement Center, School of Pharmaceutical Science, Xiamen University, Xiamen, 361102, China
| | - Qiwei Guo
- Department of Central Laboratory, Department of Obstetrics and Gynecology, Fujian Key Clinical Specialty of Laboratory Medicine, Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen, 361003, China
| | - Zhaogang Dong
- Quantum Innovation Centre, Agency for Science Technology and Research, 2 Fusionopolis Way, Innovis #08-03, Singapore, 138634, Republic of Singapore
| | - Jinfeng Zhu
- Institute of Electromagnetics and Acoustics and Key Laboratory of Electromagnetic Wave Science and Detection Technology, Xiamen University, Xiamen, 361100, China; Xiamen Dili-Chip Technology Co. Ltd., Xiamen, 361000, China; School of Electronic Science and Engineering, National Model Microelectronics College, Xiamen University, Xiamen, 361100, China.
| |
Collapse
|
2
|
Wen Z, Zhang W, Wu W. The latest applications of exosome-mediated drug delivery in anticancer therapies. Colloids Surf B Biointerfaces 2025; 249:114500. [PMID: 39799609 DOI: 10.1016/j.colsurfb.2025.114500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 12/29/2024] [Accepted: 01/05/2025] [Indexed: 01/15/2025]
Abstract
In recent years, the significant role of anticancer drugs in cancer treatment has garnered considerable attention. However, the application of these drugs is largely limited by their short half-life in blood circulation, low cellular uptake efficiency, and off-target effects. Exosomes, which serve as crucial messengers in intercellular communication, exhibit unique advantages in molecular delivery compared to traditional synthetic carriers, thereby offering new possibilities for modern drug delivery systems. Exosomes possess organotropic functions and are naturally produced by cells, making them promising candidates for natural drug delivery systems with organotropic properties and minimal side effects. These naturally derived carriers can achieve stable, efficient, and selective delivery of anticancer drugs, thereby enhancing the efficacy and potential of anticancer agents in cancer immunotherapy. This review provides a concise overview of the unique characteristics of exosomes related to anticancer drug delivery, strategies for utilizing exosomes as carriers in cancer therapy, and the latest advancements in the field.
Collapse
Affiliation(s)
- Zhiwei Wen
- School of Pharmacy, Guilin Medical University, Guilin 541199, China
| | - Wei Zhang
- School of Pharmacy, Guilin Medical University, Guilin 541199, China
| | - Wei Wu
- School of Pharmacy, Guilin Medical University, Guilin 541199, China; Guangxi Key Laboratory of Drug Discovery and Optimization, School of Pharmacy, Guilin Medical University, Guilin 541199, China.
| |
Collapse
|
3
|
Yadav K, Sahu KK, Sucheta, Minz S, Pradhan M. Unlocking exosome therapeutics: The critical role of pharmacokinetics in clinical applications. Tissue Cell 2025; 93:102749. [PMID: 39904192 DOI: 10.1016/j.tice.2025.102749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 01/10/2025] [Accepted: 01/15/2025] [Indexed: 02/06/2025]
Abstract
Exosomes are microscopic vesicles released by cells that transport various biological materials and play a vital role in intercellular communication. When they are engineered, they serve as efficient delivery systems for therapeutic agents, making it possible to precisely deliver active pharmaceutical ingredients to organs, tissues, and cells. Exosomes' pharmacokinetics, or how they are transported and metabolized inside the body, is affected by several factors, including their source of origination and the proteins in their cell membranes. The pharmacokinetics and mobility of both native and modified exosomes are being observed in living organisms using advanced imaging modalities such as in vitro-in vivo simulation, magnetic resonance imaging, and positron emission tomography. Establishing comprehensive criteria for the investigation of exosomal pharmacokinetic is essential, given its increasing significance in both therapy and diagnostics. To obtain a thorough understanding of exosome intake, distribution, metabolism, and excretion, molecular imaging methods are crucial. The development of industrial processes and therapeutic applications depends on the precise measurement of exosome concentration in biological samples. To ensure a seamless incorporation of exosomes into clinical practice, as their role in therapeutics grows, it is imperative to conduct a complete assessment of their pharmacokinetics. This review provides a brief on how exosome-based research is evolving and the need for pharmacokinetic consideration to realize the full potential of these promising new therapeutic approaches.
Collapse
Affiliation(s)
- Krishna Yadav
- Rungta College of Pharmaceutical Sciences and Research, Kohka Road, Kurud, Bhilai, Chhattisgarh 491024, India
| | - Kantrol Kumar Sahu
- Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh 281406, India
| | - Sucheta
- School of Medical and Allied Sciences, K. R. Mangalam University, Gurugram, Haryana 11 122103, India
| | - Sunita Minz
- Department of Pharmacy, Indira Gandhi National Tribal University, Amarkantak, India
| | | |
Collapse
|
4
|
Li Y, Chen L, Chen Y, Shi H, Yu S, Funmilayo A, Wu C, Wang C, Deng Y. Exosome-decorated bio-heterojunctions reduce heat and ROS transfer distance for boosted antibacterial and tumor therapy. Biomaterials 2025; 315:122921. [PMID: 39467398 DOI: 10.1016/j.biomaterials.2024.122921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 10/17/2024] [Accepted: 10/24/2024] [Indexed: 10/30/2024]
Abstract
Photothermal and photodynamic therapies represent effective modalities for combatting bacteria and tumor cells. However, therapeutic outcomes are constrained by limitations related to the heat and reactive oxygen species (ROS) transfer distance from photosensitizers to targets. To address this issue, we have devised and developed exosome-decorated bio-heterojunctions (E-bioHJ) consisted of MXene (Ti3C2), liquid metal (LM) and exosomes sourced from CT26 cells to enhance the phototherapeutic consequences. Engineering E-bioHJ enhances phototherapeutic effect in antibacterial and anti-tumor treatment, which is ascribed to reducing transfer distance of the heat and ROS. When adorned with exosomes, E-bioHJ is targetedly delivered into the cytoplasm of tumor cells to generate amount heat and ROS under 808 nm near-infrared radiation, which further induces mitochondrial dysfunction and apoptosis/necroptosis. As envisaged, this study presents a novel tactic to enhance the antibacterial and anti-tumor efficacy of biomaterials through reducing the heat and ROS delivery travel distance.
Collapse
Affiliation(s)
- Yanni Li
- West China Hospital, School of Chemical Engineering, Sichuan University, Chengdu, 610041, China; Department of Gastroenterology and Hepatology, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Lin Chen
- West China Hospital, School of Chemical Engineering, Sichuan University, Chengdu, 610041, China
| | - Yonghao Chen
- West China Hospital, School of Chemical Engineering, Sichuan University, Chengdu, 610041, China; Department of Gastroenterology and Hepatology, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Hongxing Shi
- West China Hospital, School of Chemical Engineering, Sichuan University, Chengdu, 610041, China
| | - Sheng Yu
- The School of Mechanical and Materials Engineering, Washington State University, Pullman, WA-99164, USA
| | - Adeleye Funmilayo
- The School of Mechanical and Materials Engineering, Washington State University, Pullman, WA-99164, USA
| | - Chao Wu
- Department of Orthopedics, Digital Medical Center, Zigong Fourth People's Hospital, Zigong, 643000, China
| | - Chunhui Wang
- Department of Gastroenterology and Hepatology, West China Hospital of Sichuan University, Chengdu, 610041, China; Pancreatitis Center, West China Hospital of Sichuan University, Chengdu, 610041, China.
| | - Yi Deng
- West China Hospital, School of Chemical Engineering, Sichuan University, Chengdu, 610041, China; State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, 610065, China; Department of Mechanical Engineering, The University of Hong Kong, Hong Kong, 999077, China.
| |
Collapse
|
5
|
Jiang Y, Cao Y, Yao Y, Zhang D, Wang Y. Chitosan and hyaluronic acid in breast cancer treatment: Anticancer efficacy and nanoparticle and hydrogel development. Int J Biol Macromol 2025; 301:140144. [PMID: 39848359 DOI: 10.1016/j.ijbiomac.2025.140144] [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: 12/09/2024] [Revised: 01/09/2025] [Accepted: 01/20/2025] [Indexed: 01/25/2025]
Abstract
The pervasive global health concern of breast cancer necessitates the development of innovative therapeutic interventions to enhance efficacy and mitigate adverse effects. Chitosan and hyaluronic acid, recognized for their biocompatibility and biodegradability, present compelling options for the novel drug delivery systems and therapeutic platforms in the context of breast cancer management. This review will delineate the distinctive attributes of chitosan and hyaluronic acid, encompassing their inherent anticancer properties, targeting capabilities, and suitability for chemical modifications along with nanoparticle development. These characteristics render them exceptionally well-suited for the fabrication of nanoparticles and hydrogels. The intrinsic anticancer potential of chitosan, in conjunction with its mucoadhesive properties, and the robust binding affinity of hyaluronic acid to CD44 receptors, facilitate specific drug delivery to the malignant cells, thus circumventing the limitations inherent in traditional treatment modalities such as chemotherapy. The incorporation of these materials into nanocarriers allows for the co-delivery of therapeutic agents, thereby potentiating synergistic effects, while hydrogel systems provide localized, controlled drug release and facilitate tissue regeneration. An analysis of advancements in their synthesis, functionalization, and application is presented, while also acknowledging challenges pertaining to scalability and clinical translation.
Collapse
Affiliation(s)
- Yanlin Jiang
- Department of Breast and Thyroid Surgery, the Affiliated Zhongshan Hospital of Dalian University, China
| | - Yu Cao
- Department of Surgical Oncology and Breast Surgery, The First Hospital of China Medical University, Shenyang 110001, China
| | - Yiqun Yao
- Department of Breast and Thyroid Surgery, the Affiliated Zhongshan Hospital of Dalian University, China
| | - Dianlong Zhang
- Department of Breast and Thyroid Surgery, the Affiliated Zhongshan Hospital of Dalian University, China.
| | - Yuying Wang
- Department of Breast Surgery, The Cancer Hospital of China Medical University Liaoning Cancer Hospital & Institute, China.
| |
Collapse
|
6
|
Zhang X, Ma L, Liu X, Zhou X, Wang A, Lai Y, Zhang J, Li Y, Chen S. Sustained release of miR-21 carried by mesenchymal stem cell-derived exosomes from GelMA microspheres inhibits ovarian granulosa cell apoptosis in premature ovarian insufficiency. Mater Today Bio 2025; 31:101469. [PMID: 39906205 PMCID: PMC11790500 DOI: 10.1016/j.mtbio.2025.101469] [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: 04/18/2024] [Revised: 01/04/2025] [Accepted: 01/07/2025] [Indexed: 02/06/2025] Open
Abstract
Background Premature ovarian insufficiency (POI) refers to the severe decline or failure of ovarian function in women younger than 40 years of age. It is a serious hazard to women's physical and mental health, but current treatment options are limited. Mesenchymal stem cell-derived exosomes (MSC-Exo) exhibit promising potential as a therapeutic approach for POI. However, their clinical application is hindered by their instability and low long-term retention rate in vivo. Methods and results In this study, miR-21 was identified as the predominant miRNA with low-expression in follicular fluid exosomes of POI patients and was shown to possess antiapoptotic activity. Next, we loaded miR-21 agomir to MSC-Exo to form Agomir21-Exo, which significantly reversed the apoptosis of granulosa cells in vitro. Moreover, we successfully developed GelMA hydrogel microspheres for encapsulating Agomir21-Exo through microfluidic technology, named GelMA-Ag21Exo, which had good injectability and significantly enhanced the stability and long-term retention of Agomir21-Exo in mice through sustained release. The release of Agomir21-Exo from GelMA-Ag21Exo notably alleviated the apoptosis of ovarian granulosa cells and improved the ovarian reserve and fertility in POI mice. Conclusion Our findings illustrate that activating miR-21 through Agomir21-Exo could improve the function of ovarian granulosa cells. The GelMA-Ag21Exo enhanced the exosome-based therapeutic efficacy of the Agomir21-Exo in vivo. These findings provide a novel and promising treatment strategy for POI patients.
Collapse
Affiliation(s)
| | | | | | - Xingyu Zhou
- Center for Reproductive Medicine, Department of Gynecology and Obstetrics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Ao Wang
- Center for Reproductive Medicine, Department of Gynecology and Obstetrics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Yunhui Lai
- Center for Reproductive Medicine, Department of Gynecology and Obstetrics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Jun Zhang
- Center for Reproductive Medicine, Department of Gynecology and Obstetrics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Ying Li
- Center for Reproductive Medicine, Department of Gynecology and Obstetrics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Shiling Chen
- Center for Reproductive Medicine, Department of Gynecology and Obstetrics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| |
Collapse
|
7
|
Wu P, Wang Z, Sun Y, Cheng Z, Wang M, Wang B. Extracellular vesicles: a new frontier in diagnosing and treating graft-versus-host disease after allogeneic hematopoietic cell transplantation. J Nanobiotechnology 2025; 23:251. [PMID: 40133949 PMCID: PMC11938667 DOI: 10.1186/s12951-025-03297-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 03/04/2025] [Indexed: 03/27/2025] Open
Abstract
Graft-versus-host disease (GvHD) is a prevalent complication following allogeneic hematopoietic stem cell transplantation (HSCT) and is characterized by relatively high morbidity and mortality rates. GvHD can result in extensive systemic damage in patients following allogeneic HSCT (allo-HSCT), with the skin, gastrointestinal tract, and liver frequently being the primary target organs affected. The severe manifestations of acute intestinal GvHD often indicate a poor prognosis for patients after allo-HSCT. Endoscopy and histopathological evaluation remain employed to diagnose GvHD, and auxiliary examinations exclude differential diagnoses. Currently, reliable serum biomarkers for the diagnosis and differential diagnosis of GvHD are scarce. As an essential part of standard transplant protocols, early application of immunosuppressive drugs effectively prevents GvHD. Among them, steroids represent first-line therapeutic agents, and the JAK2 inhibitor ruxolitinib represents the second-line therapeutic agent. Currently, no efficacious treatment modality exists for steroid-resistant aGvHD. Therefore, the diagnosis and treatment of GvHD still face significant medical demands. Extracellular vesicles (EVs) are nanometer to micrometer-scale biomembrane vesicles containing various bioactive components, such as proteins, nucleotides, and metabolites. Distinctive changes in serum-derived EV components occur in patients after allo-HSCT; Hence, EVs are expected to be potential biomarkers for diagnosing and treating GvHD. Furthermore, cell-free therapeutics characterized by EVs derived from mesenchymal stem cells (MSCs) have manifested remarkable therapeutic efficacy in preclinical models and preclinical trials of GvHD. Customized engineered EVs with fewer toxic and side effects for the combined treatment of GvHD hold broad prospects for clinical translation. This review article examines the potential value of translating EVs into clinical applications for the diagnosis and treatment of GvHD. It summarizes the latest advancements and prospects of engineered EVs applying GvHD.
Collapse
Affiliation(s)
- Peipei Wu
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Core Unit of National Clinical Research Center for Laboratory Medicine, Hefei, China
| | - Zhangfei Wang
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Core Unit of National Clinical Research Center for Laboratory Medicine, Hefei, China
| | - Yongping Sun
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Zhixiang Cheng
- Department of Blood Transfusion, The First Affiliated Hospital of Anhui Medical University, Hefei, China.
- Anhui Public Health Clinical Center, Hefei, China.
| | - Min Wang
- School of Biomedical Engineering, Research and Engineering Center of Biomedical Materials, Anhui Medical University, Hefei, China.
| | - Baolong Wang
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
- Core Unit of National Clinical Research Center for Laboratory Medicine, Hefei, China.
| |
Collapse
|
8
|
Jin JJ, Liu RH, Chen JY, Wang K, Han JY, Nie DS, Gong YQ, Lin B, Weng GX. MiR-21-5p-enriched exosomes from hiPSC-derived cardiomyocytes exhibit superior cardiac repair efficacy compared to hiPSC-derived exosomes in a murine MI model. World J Stem Cells 2025; 17:101454. [DOI: 10.4252/wjsc.v17.i3.101454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Revised: 10/15/2024] [Accepted: 01/15/2025] [Indexed: 03/21/2025] Open
Abstract
BACKGROUND Heart disease remains a leading cause of mortality worldwide, with existing treatments often failing to effectively restore damaged myocardium. Human-induced pluripotent stem cells (hiPSCs) and their derivatives offer promising therapeutic options; however, challenges such as low retention, engraftment issues, and tumorigenic risks hinder their clinical utility. Recent focus has shifted to exosomes (exos) - nanoscale vesicles that facilitate intercellular communication - as a safer and more versatile alternative. Understanding the specific mechanisms and comparative efficacy of exos from hiPSCs vs hiPSC-derived cardiomyocytes (hiPSC-CMs) is crucial for advancing cardiac repair therapies.
AIM To evaluate and compare the therapeutic efficacy of exos secreted by hiPSCs and hiPSC-CMs in cardiac repair, and to elucidate the role of microRNA 21-5p (miR-21-5p) in the observed effects.
METHODS We differentiated hiPSCs into CMs using small molecule methods and characterized the cells and their exos.
RESULTS Our findings indicate that hiPSC-CMs and their exos enhanced cardiac function, reduced infarct size, and decreased myocardial fibrosis in a murine myocardial infarction model. Notably, hiPSC-CM exos outperformed hiPSC-CM cell therapy, showing improved ejection fraction and reduced apoptosis. We identified miR-21-5p, a microRNA in hiPSC-CM exos, as crucial for CM survival. Exos with miR-21-5p were absorbed by AC16 cells, suggesting a mechanism for their cytoprotective effects.
CONCLUSION Overall, hiPSC-CM exos could serve as a potent therapeutic agent for myocardial repair, laying the groundwork for future research into exos as a treatment for ischemic heart disease.
Collapse
Affiliation(s)
- Jing-Jun Jin
- Fujian Key Laboratory of Medical Analysis, Fujian Academy of Medical Sciences, Fuzhou 350001, Fujian Province, China
| | - Rong-Hua Liu
- Fujian Key Laboratory of Medical Analysis, Fujian Academy of Medical Sciences, Fuzhou 350001, Fujian Province, China
| | - Jin-Yan Chen
- Fujian Key Laboratory of Medical Analysis, Fujian Academy of Medical Sciences, Fuzhou 350001, Fujian Province, China
| | - Kun Wang
- Fujian Key Laboratory of Medical Analysis, Fujian Academy of Medical Sciences, Fuzhou 350001, Fujian Province, China
| | - Jun-Yong Han
- Fujian Key Laboratory of Medical Analysis, Fujian Academy of Medical Sciences, Fuzhou 350001, Fujian Province, China
| | - Dao-Shun Nie
- Shengli Clinical Medical College, Fujian Medical University, Fuzhou 350001, Fujian Province, China
| | - Yu-Qing Gong
- Shengli Clinical Medical College, Fujian Medical University, Fuzhou 350001, Fujian Province, China
| | - Bin Lin
- Shengli Clinical Medical College, Fujian Medical University, Fuzhou 350001, Fujian Province, China
| | - Guo-Xing Weng
- Shengli Clinical Medical College, Fujian Medical University, Fuzhou 350001, Fujian Province, China
| |
Collapse
|
9
|
Semeradtova A, Liegertova M, Herma R, Capkova M, Brignole C, Del Zotto G. Extracellular vesicles in cancer´s communication: messages we can read and how to answer. Mol Cancer 2025; 24:86. [PMID: 40108630 PMCID: PMC11921637 DOI: 10.1186/s12943-025-02282-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Accepted: 02/24/2025] [Indexed: 03/22/2025] Open
Abstract
Extracellular vesicles (EVs) are emerging as critical mediators of intercellular communication in the tumor microenvironment (TME), profoundly influencing cancer progression. These nano-sized vesicles, released by both tumor and stromal cells, carry a diverse cargo of proteins, nucleic acids, and lipids, reflecting the dynamic cellular landscape and mediating intricate interactions between cells. This review provides a comprehensive overview of the biogenesis, composition, and functional roles of EVs in cancer, highlighting their significance in both basic research and clinical applications. We discuss how cancer cells manipulate EV biogenesis pathways to produce vesicles enriched with pro-tumorigenic molecules, explore the specific contributions of EVs to key hallmarks of cancer, such as angiogenesis, metastasis, and immune evasion, emphasizing their role in shaping TME and driving therapeutic resistance. Concurrently, we submit recent knowledge on how the cargo of EVs can serve as a valuable source of biomarkers for minimally invasive liquid biopsies, and its therapeutic potential, particularly as targeted drug delivery vehicles and immunomodulatory agents, showcasing their promise for enhancing the efficacy and safety of cancer treatments. By deciphering the intricate messages carried by EVs, we can gain a deeper understanding of cancer biology and develop more effective strategies for early detection, targeted therapy, and immunotherapy, paving the way for a new era of personalized and precise cancer medicine with the potential to significantly improve patient outcomes.
Collapse
Affiliation(s)
- Alena Semeradtova
- Institute of Photonics and Electronics of the CAS, Chaberská 1014/57, Prague, 182 51, Czech Republic.
| | - Michaela Liegertova
- Centre for Nanomaterials and Biotechnology, Faculty of Science, Jan Evangelista Purkyně University in Ústí Nad Labem, Pasteurova 3632/15, Ústí Nad Labem, 40096, Czech Republic
| | - Regina Herma
- Centre for Nanomaterials and Biotechnology, Faculty of Science, Jan Evangelista Purkyně University in Ústí Nad Labem, Pasteurova 3632/15, Ústí Nad Labem, 40096, Czech Republic
| | - Magdalena Capkova
- Institute of Photonics and Electronics of the CAS, Chaberská 1014/57, Prague, 182 51, Czech Republic
| | - Chiara Brignole
- Laboratory of Experimental Therapies in Oncology, IRCCS Istituto Giannina Gaslini, Via G. Gaslini 5, 16147, Genoa, Italy.
| | - Genny Del Zotto
- Core Facilities, Department of Research and Diagnostics, IRCCS Istituto Giannina Gaslini, 16147, Genoa, Italy.
| |
Collapse
|
10
|
Alfawaz Altamimi AS, Arockia Babu M, Afzal M, Bishoyi AK, Roopashree R, Saini S, Sharma RSK, Pathak PK, Chauhan AS, Goyal K, Ali H, Khan NH, Balaraman AK. Exosomes derived from natural killer cells: transforming immunotherapy for aggressive breast cancer. Med Oncol 2025; 42:114. [PMID: 40100465 DOI: 10.1007/s12032-025-02647-y] [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: 12/01/2024] [Accepted: 02/24/2025] [Indexed: 03/20/2025]
Abstract
Natural killer cell-derived exosomes (NK-Exos) hold great promise as immune modulators and immunotherapeutics against cancer due to their intrinsically latent anti-tumor effects. They use these nanosized vesicles to deliver cytotoxic molecules, such as perforin, granzymes, and miRNAs, directly to cancer cells to kill them, avoiding immune suppression. NK-Exos has particular efficacy for treating aggressive breast cancer by modulating the TME to activate the immune response and suppress immunosuppressive factors. Bioengineering advances have extended the therapeutic potential of NK-Exos, which permits precise tumor cell targeting and efficient delivery of therapeutic payloads, including small RNAs and chemotherapeutic agents. In engineered NK-Exos, sensitization of cancer cells to apoptosis, reduction of tumor growth, and resistance to drugs have been demonstrated to be highly effective. When combined, NK-Exos synergizes with radiotherapy, chemotherapy, or checkpoint inhibitors, enhancing therapeutic efficacy, and minimizing systemic toxicity. This review emphasizes the critical role of NK-Exos in breast cancer treatment, their integration into combination therapies, and the need for further research to overcome existing limitations and fully realize their clinical potential.
Collapse
Affiliation(s)
| | - M Arockia Babu
- Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh, India
| | - Muhammad Afzal
- Department of Pharmaceutical Sciences, Pharmacy Program, Batterjee Medical College, P.O. Box 6231, 21442, Jeddah, Saudi Arabia
| | - Ashok Kumar Bishoyi
- Department of Microbiology, Faculty of Science, Marwadi University Research Center, Marwadi University, Rajkot, Gujarat, 360003, India
| | - R Roopashree
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Suman Saini
- Department of Chemistry, Chandigarh Engineering College, Chandigarh Group of Colleges-Jhanjeri, Mohali, Punjab, 140307, India
| | - R S K Sharma
- Department of Chemistry, Raghu Engineering College, Visakhapatnam, Andhra Pradesh, 531162, India
| | - Piyus Kumar Pathak
- Department of Applied Sciences-Chemistry, NIMS Institute of Engineering & Technology, NIMS University Rajasthan, Jaipur, India
| | - Ashish Singh Chauhan
- Division of Research and Innovation, Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, Uttarakhand, India
| | - Kavita Goyal
- Department of Biotechnology, Graphic Era (Deemed to be University), Clement Town, Dehradun, 248002, India
| | - Haider Ali
- Faculty of Medicine, Ala-Too International University, Bishkek, Kyrgyz Republic
| | - Nawaid Hussain Khan
- Faculty of Medicine, Ala-Too International University, Bishkek, Kyrgyz Republic
| | - Ashok Kumar Balaraman
- Research and Enterprise, University of Cyberjaya, Persiaran Bestari, Cyber 11, 63000, Cyberjaya, Selangor, Malaysia.
| |
Collapse
|
11
|
Li T, Zhou Y, Wang H, Wang J, Lu R. Leveraging engineered yeast small extracellular vesicles serve as multifunctional platforms for effectively loading methyl salicylate through the "esterase-responsive active loading" strategy. Eur J Pharm Biopharm 2025:114696. [PMID: 40113048 DOI: 10.1016/j.ejpb.2025.114696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 03/11/2025] [Accepted: 03/13/2025] [Indexed: 03/22/2025]
Abstract
Small extracellular vesicles (sEVs) are a promising vehicle for drug delivery because of their good biocompatibility and nontoxicity. The drug loading and encapsulation efficiencies of them are not satisfactory. This is especially the case when drugs are loaded by co-incubation. In this situation, as the difference in drug concentration between the inside and outside of the membrane of ordinary sEVs decreases, the drugs cannot diffuse efficiently into the inside of the vesicles. As a result, the drug loading efficiency is low. In this study, engineered yeast-derived small extracellular vesicles derived from Pichia pastoris X33 (XPP-sEVs) engineered with carboxylesterase 1 (CES1) were constructed using the "esterase-responsive active loading" method, which is based on the concept of prodrug design and guided by the strategy of immobilized enzymes, to improve the loading efficiency of methyl salicylate (MS) to about twice as much. This was achieved by engineering the CES1-contained small extracellular vesicles to catalyze the esterase hydrolysis reaction of MS to establish a continuous MS transmembrane concentration gradient for efficient loading of the active drugs, including methyl salicylate and its hydrolyzed active product salicylic acid. The results showed that the enzyme activity of the CES1-sEVs group finally reached 7.88 ± 0.43 U/mL, and the drug loading efficiency was about doubled. The results of drug release from the engineered extracellular vesicles showed that the release of the drug reached equilibrium around 100 min-2 h, during which there was no sudden release of the MS, and the final amount of the drug released could be increased by 12.34 % compared with the emulsion dosage form of the MS. Overall, the CES1-sEVs prepared in this study significantly improved the drug-loading efficiency of MS without affecting the anti-inflammatory activity of MS. The MS-CES1-sEVs prepared in this study are non-toxic and have a bright application prospect in the treatment of skin inflammation.
Collapse
Affiliation(s)
- Tianhao Li
- Marine College, Shandong University, No. 180 Wenhua West Road, Weihai 264209, China.
| | - Yun Zhou
- Marine College, Shandong University, No. 180 Wenhua West Road, Weihai 264209, China.
| | - Haoran Wang
- Weihai Neoland Biosciences Co., Ltd. C-301, Torch Innovative Imbark Base, No. 213-2 Huoju Road, Weihai 264209 Shandong, China.
| | - Junfeng Wang
- Weihai Neoland Biosciences Co., Ltd. C-301, Torch Innovative Imbark Base, No. 213-2 Huoju Road, Weihai 264209 Shandong, China.
| | - Rong Lu
- Marine College, Shandong University, No. 180 Wenhua West Road, Weihai 264209, China; Weihai Neoland Biosciences Co., Ltd. C-301, Torch Innovative Imbark Base, No. 213-2 Huoju Road, Weihai 264209 Shandong, China.
| |
Collapse
|
12
|
Madhan S, Dhar R, Devi A. Clinical Impact of Exosome Chemistry in Cancer. ACS APPLIED BIO MATERIALS 2025; 8:1862-1876. [PMID: 39936581 DOI: 10.1021/acsabm.4c01920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/13/2025]
Abstract
As we progress into the 21st century, cancer stands as one of the most dreaded diseases. With approximately one in every four individuals facing a lifetime risk of developing cancer, cancer remains one of the most serious health challenges worldwide. Its multifaceted nature makes it an arduous and tricky problem to diagnose and treat. Over the years, researchers have explored plenty of approaches and avenues to improve cancer management. One notable strategy includes the study of extracellular vesicles (EVs) as potential biomarkers and therapeutics. Among these EVs, exosomes have emerged as particularly promising candidates due to their unique characteristic properties and functions. They are small membrane-bound vesicles secreted by cells carrying a cargo of biomolecules such as proteins, nucleic acids, and lipids. These vesicles play crucial roles in intercellular communication, facilitating the transfer of biological information between cell-to-cell communication. Exosomes transport cargoes such as DNA, RNA, proteins, and lipids involved in cellular reprogramming and promoting cancer. In this review, we explore the molecular composition of exosomes, significance of exosomes chemistry in cancer development, and its theranostic application as well as exosomes research complications and solutions.
Collapse
Affiliation(s)
- Shrishti Madhan
- Cancer and Stem Cell Biology Laboratory, Department of Genetic Engineering, SRM Institute of Science and Technology, Kattankulathur, Chengalpattu District, Tamil Nadu-603203, India
| | - Rajib Dhar
- Cancer and Stem Cell Biology Laboratory, Department of Genetic Engineering, SRM Institute of Science and Technology, Kattankulathur, Chengalpattu District, Tamil Nadu-603203, India
| | - Arikketh Devi
- Cancer and Stem Cell Biology Laboratory, Department of Genetic Engineering, SRM Institute of Science and Technology, Kattankulathur, Chengalpattu District, Tamil Nadu-603203, India
| |
Collapse
|
13
|
Du W, Chen C, Liu Y, Quan H, Xu M, Liu J, Song P, Fang Z, Yue Z, Xu H, Ling Y, Duan J, He F, Wang L. A combined "eat me/don't eat me" strategy based on exosome for acute liver injury treatment. Cell Rep Med 2025:102033. [PMID: 40120577 DOI: 10.1016/j.xcrm.2025.102033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 12/09/2024] [Accepted: 02/24/2025] [Indexed: 03/25/2025]
Abstract
Drug-induced liver injury (DILI) involves multifaceted pathogenesis, necessitating effective therapeutic strategies. Wnt2, secreted by liver sinusoidal endothelial cell (LSEC), activates the Wnt/β-catenin signaling pathway to promote hepatocyte proliferation after injury. To address the dual challenges of targeted delivery and phagocytosis evasion, we develop a combined "eat me/don't eat me" strategy. RLTRKRGLK (RLTR) peptide-functionalized exosomes are engineered by inserting DMPE-PEG2000-CRLTRKRGLK into the lipid membrane of exosome derived from bEnd.3 cell. Surface-displayed RLTR mediates exosomal enrichment in LSEC, while CD47 engineering reduces macrophage clearance via "don't eat me" signaling. Then, lentiviral transfection enables stable encapsulation of functional Wnt2 mRNA into ExoCD47 (designated Wnt2@ExoCD47). In both acetaminophen (APAP) and dimethylnitrosamine (DMN)-induced murine liver injury models, RLTR-Wnt2@ExoCD47 demonstrates LSEC-specific targeting and significant hepatoprotection. This engineered exosome platform provides a therapeutic strategy for DILI.
Collapse
Affiliation(s)
- Wei Du
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Chen Chen
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - YingYing Liu
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Huiyi Quan
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Ming Xu
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - JingJing Liu
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Ping Song
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - ZhiQiang Fang
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - ZhenSheng Yue
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Hao Xu
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - YuWei Ling
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - JuanLi Duan
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China.
| | - Fei He
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China.
| | - Lin Wang
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China.
| |
Collapse
|
14
|
Kim D, Choi J, Jin D, Xu E, Lee J, Byun J, Oh YK. Hybrid lipid nanoparticles with tumor antigen-primed dendritic cell membranes for post-surgical tumor immunotherapy. J Control Release 2025; 379:537-548. [PMID: 39800239 DOI: 10.1016/j.jconrel.2025.01.027] [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: 09/15/2024] [Revised: 12/22/2024] [Accepted: 01/09/2025] [Indexed: 01/15/2025]
Abstract
Post-surgical tumor recurrence poses a major challenge in cancer treatment due to residual tumor cells and surgery-induced immunosuppression. Here, we developed hybrid nanoparticles, termed T-DCNPs, designed to promote antigen-specific activation of cytotoxic CD8+ T cells while concurrently inhibiting immunosuppressive pathways within the tumor microenvironment. T-DCNPs were formulated by co-extruding lipid nanoparticles containing a transforming growth factor β inhibitor with dendritic cells that were pre-treated with autologous neoantigens derived from surgically excised tumors. By using whole tumor antigens rather than specific peptides, T-DCNPs effectively overcame tumor heterogeneity and elicited a robust, targeted immune response. In vitro studies showed that T-DCNPs enhanced CD8+ T cell proliferation and reduced programmed death-1 (PD-1) expression, leading to increased antitumor cytotoxicity. In vivo experiments, involving intratumoral injections of T-DCNPs in distant tumor and post-surgical melanoma models, demonstrated a significant reduction in distant tumor growth, decreased recurrence rates, and extended survival compared to control groups. Flow cytometry and immunohistochemistry analyses further confirmed the enhanced infiltration of activated CD8+ T cells and a marked reduction in immunosuppressive markers, including PD-1 and Foxp3, within the treated tumors. These results suggest that T-DCNPs, through the dual mechanisms of tumor antigen-specific T cell activation and immune modulation, offer a promising strategy to prevent tumor recurrence following surgery and could potentially improve the efficacy of postoperative cancer immunotherapy.
Collapse
Affiliation(s)
- Dongyoon Kim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Jaehyun Choi
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Dongun Jin
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Enzhen Xu
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Jaiwoo Lee
- College of Pharmacy, Korea University, Sejong 30019, Republic of Korea.
| | - Junho Byun
- College of Pharmacy, Sookmyung Women's University, Seoul 04310, Republic of Korea.
| | - Yu-Kyoung Oh
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea.
| |
Collapse
|
15
|
Youssef E, Palmer D, Fletcher B, Vaughn R. Exosomes in Precision Oncology and Beyond: From Bench to Bedside in Diagnostics and Therapeutics. Cancers (Basel) 2025; 17:940. [PMID: 40149276 DOI: 10.3390/cancers17060940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 02/28/2025] [Accepted: 03/07/2025] [Indexed: 03/29/2025] Open
Abstract
Exosomes have emerged as pivotal players in precision oncology, offering innovative solutions to longstanding challenges such as metastasis, therapeutic resistance, and immune evasion. These nanoscale extracellular vesicles facilitate intercellular communication by transferring bioactive molecules that mirror the biological state of their parent cells, positioning them as transformative tools for cancer diagnostics and therapeutics. Recent advancements in exosome engineering, artificial intelligence (AI)-driven analytics, and isolation technologies are breaking barriers in scalability, reproducibility, and clinical application. Bioengineered exosomes are being leveraged for CRISPR-Cas9 delivery, while AI models are enhancing biomarker discovery and liquid biopsy accuracy. Despite these advancements, key obstacles such as heterogeneity in exosome populations and the lack of standardized isolation protocols persist. This review synthesizes pioneering research on exosome biology, molecular engineering, and clinical translation, emphasizing their dual roles as both mediators of tumor progression and tools for intervention. It also explores emerging areas, including microbiome-exosome interactions and the integration of machine learning in exosome-based precision medicine. By bridging innovation with translational strategies, this work charts a forward-looking path for integrating exosomes into next-generation cancer care, setting it apart as a comprehensive guide to overcoming clinical and technological hurdles in this rapidly evolving field.
Collapse
|
16
|
Aswani BS, Sajeev A, Hegde M, Mishra A, Abbas M, Vayalpurayil T, Sethi G, Kunnumakkara AB. Exosomal dynamics: Bridging the gap between cellular senescence and cancer therapy. Mech Ageing Dev 2025; 225:112045. [PMID: 40074065 DOI: 10.1016/j.mad.2025.112045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 03/01/2025] [Accepted: 03/03/2025] [Indexed: 03/14/2025]
Abstract
Cancer remains one of the most devastating diseases, severely affecting public health and contributing to economic instability. Researchers worldwide are dedicated to developing effective therapeutics to target cancer cells. One promising strategy involves inducing cellular senescence, a complex state in which cells exit the cell cycle. Senescence has profound effects on both physiological and pathological processes, influencing cellular systems through secreted factors that affect surrounding and distant cells. Among these factors are exosomes, small extracellular vesicles that play crucial roles in cellular communication, development, and defense, and can contribute to pathological conditions. Recently, there has been increasing interest in engineering exosomes as precise drug delivery vehicles, capable of targeting specific cells or intracellular components. Studies have emphasized the significant role of exosomes from senescent cells in cancer progression and therapy. Notably, chemotherapeutic agents can alter the tumor microenvironment, induce senescence, and trigger immune responses through exosome-mediated cargo transfer. This review explores the intricate relationship between cellular senescence, exosomes, and cancer, examining how different therapeutics can eliminate cancer cells or promote drug resistance. It also investigates the molecular mechanisms and signaling pathways driving these processes, highlighting current challenges and proposing future perspectives to uncover new therapeutic strategies for cancer treatment.
Collapse
Affiliation(s)
- Babu Santha Aswani
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, Assam 781039, India
| | - Anjana Sajeev
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, Assam 781039, India
| | - Mangala Hegde
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, Assam 781039, India
| | - Anamika Mishra
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, Assam 781039, India
| | - Mohamed Abbas
- Electrical Engineering Department, College of Engineering, King Khalid University, Abha 61421, Saudi Arabia
| | - Thafasalijyas Vayalpurayil
- Electrical Engineering Department, College of Engineering, King Khalid University, Abha 61421, Saudi Arabia
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 117600, Singapore; NUS Center for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, 117699, Singapore.
| | - Ajaikumar B Kunnumakkara
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, Assam 781039, India.
| |
Collapse
|
17
|
Yu B, Wang L, Mao Y, Hu X, Lu Y, He J, Yuan X, Zhang M, Chen Z. Research progress on small extracellular vesicles in diabetic nephropathy. Front Cell Dev Biol 2025; 13:1535249. [PMID: 40109365 PMCID: PMC11920185 DOI: 10.3389/fcell.2025.1535249] [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: 11/27/2024] [Accepted: 02/06/2025] [Indexed: 03/22/2025] Open
Abstract
Virtually all cell types are capable of secreting small extracellular vesicles (sEV), which can be internalized by recipient cells, thereby serving as vehicles for intercellular communication. The cargoes of these vesicles, such as microRNAs, circular RNAs, proteins, and lipids, play significant roles in both normal cellular functions and the pathogenesis of various diseases. Diabetic Nephropathy (DN), a complication arising from diabetes, is expected to contribute to a 54% increase in the global diabetic population between 2015 and 2030, leading to substantial economic burdens on individuals and healthcare systems. sEVs, as promising biomarkers, demonstrate diverse mechanistic responses in different types of Diabetic Kidney Disease (DKD). They also hold advantages in the early prediction of renal damage. This article reviews the functional mechanisms of sEVs in DKD and their potential as therapeutic targets and biomarkers.
Collapse
Affiliation(s)
- Bingqing Yu
- Department of Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
- College of Medical Technology, Gannan Medical University, Ganzhou, Jiangxi, China
| | - Lanfeng Wang
- Department of Nephrology, First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Yiping Mao
- Department of Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
- College of Medical Technology, Gannan Medical University, Ganzhou, Jiangxi, China
| | - Xinyi Hu
- Department of Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
- College of Medical Technology, Gannan Medical University, Ganzhou, Jiangxi, China
| | - Yukang Lu
- Department of Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
- First Clinical Medical College, Gannan Medical University, Ganzhou, Jiangxi, China
| | - Jiahui He
- Department of Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
- College of Medical Technology, Gannan Medical University, Ganzhou, Jiangxi, China
| | - Xiaoying Yuan
- First Clinical Medical College, Gannan Medical University, Ganzhou, Jiangxi, China
| | - Man Zhang
- First Clinical Medical College, Gannan Medical University, Ganzhou, Jiangxi, China
| | - Zhiping Chen
- Department of Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
- College of Medical Technology, Gannan Medical University, Ganzhou, Jiangxi, China
- First Clinical Medical College, Gannan Medical University, Ganzhou, Jiangxi, China
| |
Collapse
|
18
|
Wang J, Shao F, Yu QX, Ye L, Wusiman D, Wu R, Tuo Z, Wang Z, Li D, Cho WC, Wei W, Feng D. The Common Hallmarks and Interconnected Pathways of Aging, Circadian Rhythms, and Cancer: Implications for Therapeutic Strategies. RESEARCH (WASHINGTON, D.C.) 2025; 8:0612. [PMID: 40046513 PMCID: PMC11880593 DOI: 10.34133/research.0612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Revised: 01/14/2025] [Accepted: 01/24/2025] [Indexed: 03/17/2025]
Abstract
The intricate relationship between cancer, circadian rhythms, and aging is increasingly recognized as a critical factor in understanding the mechanisms underlying tumorigenesis and cancer progression. Aging is a well-established primary risk factor for cancer, while disruptions in circadian rhythms are intricately associated with the tumorigenesis and progression of various tumors. Moreover, aging itself disrupts circadian rhythms, leading to physiological changes that may accelerate cancer development. Despite these connections, the specific interplay between these processes and their collective impact on cancer remains inadequately explored in the literature. In this review, we systematically explore the physiological mechanisms of circadian rhythms and their influence on cancer development. We discuss how core circadian genes impact tumor risk and prognosis, highlighting the shared hallmarks of cancer and aging such as genomic instability, cellular senescence, and chronic inflammation. Furthermore, we examine the interplay between circadian rhythms and aging, focusing on how this crosstalk contributes to tumorigenesis, tumor proliferation, and apoptosis, as well as the impact on cellular metabolism and genomic stability. By elucidating the common pathways linking aging, circadian rhythms, and cancer, this review provides new insights into the pathophysiology of cancer and identifies potential therapeutic strategies. We propose that targeting the circadian regulation of cancer hallmarks could pave the way for novel treatments, including chronotherapy and antiaging interventions, which may offer important benefits in the clinical management of cancer.
Collapse
Affiliation(s)
- Jie Wang
- Department of Urology, Institute of Urology, West China Hospital,
Sichuan University, Chengdu 610041, China
| | - Fanglin Shao
- Department of Rehabilitation,
The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Qing Xin Yu
- Department of Pathology,
Ningbo Clinical Pathology Diagnosis Center, Ningbo, Zhejiang 315211, China
- Department of Pathology,
Ningbo Medical Centre Lihuili Hospital, Ningbo, Zhejiang 315040, China
| | - Luxia Ye
- Department of Public Research Platform,
Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
| | - Dilinaer Wusiman
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN 47907, USA
- Purdue Institute for Cancer Research, Purdue University, West Lafayette, IN 47906, USA
| | - Ruicheng Wu
- Department of Urology, Institute of Urology, West China Hospital,
Sichuan University, Chengdu 610041, China
| | - Zhouting Tuo
- Department of Urological Surgery, Daping Hospital, Army Medical Center of PLA,
Army Medical University, Chongqing, China
| | - Zhipeng Wang
- Department of Urology, Sichuan Provincial People’s Hospital,
University of Electronic Science and Technology of China, Chengdu, China
| | - Dengxiong Li
- Department of Urology, Institute of Urology, West China Hospital,
Sichuan University, Chengdu 610041, China
| | - William C. Cho
- Department of Clinical Oncology,
Queen Elizabeth Hospital, Hong Kong SAR, China
| | - Wuran Wei
- Department of Urology, Institute of Urology, West China Hospital,
Sichuan University, Chengdu 610041, China
| | - Dechao Feng
- Department of Urology, Institute of Urology, West China Hospital,
Sichuan University, Chengdu 610041, China
- Division of Surgery and Interventional Science,
University College London, London W1W 7TS, UK
| |
Collapse
|
19
|
Poorva P, Mast J, Cao B, Shah MV, Pollok KE, Shen J. Killing the killers: Natural killer cell therapy targeting glioma stem cells in high-grade glioma. Mol Ther 2025:S1525-0016(25)00168-6. [PMID: 40040281 DOI: 10.1016/j.ymthe.2025.02.043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 01/22/2025] [Accepted: 02/26/2025] [Indexed: 03/06/2025] Open
Abstract
High-grade gliomas (HGGs), including glioblastoma (GBM) in adults and diffuse intrinsic pontine glioma (DIPG) in children, are among the most aggressive and deadly brain tumors. A key factor in their resilience is the presence of glioma stem cells (GSCs), which drive tumor initiation, progression, and resistance to treatment. Targeting and eradicating GSCs holds potential for curing both GBM and DIPG. Natural killer (NK) cells, as part of the innate immune system, naturally recognize and destroy malignant cells. Recent advances in NK cell-based therapies, such as chimeric antigen receptor (CAR)-NK cells, NK cell engagers, and NK cell-derived exosomes, offer promising approaches for treating GBM and DIPG, particularly by addressing the persistence of GSCs. This review highlights these advancements, explores challenges such as the blood-brain barrier and the immunosuppressive tumor microenvironment, and proposes future directions for improving and clinically advancing these NK cell-based therapies for HGGs.
Collapse
Affiliation(s)
- Poorva Poorva
- Medical Sciences Program, Indiana University School of Medicine, Bloomington, IN 47405, USA
| | - Jensen Mast
- Biochemistry Graduate Program, Indiana University, Bloomington, IN 47405, USA
| | - Bihui Cao
- Medical Sciences Program, Indiana University School of Medicine, Bloomington, IN 47405, USA
| | - Mitesh V Shah
- Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Karen E Pollok
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indianapolis, IN 46202, USA
| | - Jia Shen
- Medical Sciences Program, Indiana University School of Medicine, Bloomington, IN 47405, USA; Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indianapolis, IN 46202, USA; Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| |
Collapse
|
20
|
Teng X, Wang X, Wang Z. Mesenchymal Stromal Cell Exosome-Induced Vascular Regeneration in a PCOS Mouse Model. Reprod Sci 2025; 32:825-835. [PMID: 39407058 DOI: 10.1007/s43032-024-01720-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 09/28/2024] [Indexed: 03/03/2025]
Abstract
The efficacy of Bone Marrow Mesenchymal Stem Cell-derived Exosomes (BMSCs-Exo) in addressing the complexities of Polycystic Ovary Syndrome (PCOS) has been explored in a controlled experimental study using a DHEA-induced PCOS model in 6-8-week-old female NMRI mice. This research undertook an in vivo approach with fifteen female murine subjects to investigate the potential of BMSCs-Exo in promoting vascular regeneration and alleviating the adverse effects associated with PCOS. Through a strategic intervention, the study aimed to modulate the pathophysiological markers of oxidative stress and inflammation that are hallmark features of PCOS. Remarkably, the administration of BMSCs-Exo led to decreased CD31 expression in ovarian tissues, suggesting reduced angiogenesis and endothelial activation. Moreover, a significant reduction in pro-inflammatory cytokines and oxidative stress markers was noted, aligning closely with the metrics observed in the control group. These findings illuminate a promising therapeutic avenue utilizing BMSCs-Exo to recalibrate angiogenic, inflammatory, and oxidative stress responses in PCOS. This research not only contributes to the current understanding of PCOS management but also opens new doors for innovative clinical treatments.
Collapse
Affiliation(s)
- Xiaojing Teng
- Department of Clinical Laboratory, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, 310000, Zhejiang, China
| | - Xiaolei Wang
- Department of Clinical Laboratory, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, 310000, Zhejiang, China
| | - Zhiyi Wang
- Department of Clinical Laboratory, Hangzhou Women's Hospital (Hangzhou Maternity and Child Health Care Hospital), Hangzhou, 310008, Zhejiang, China.
| |
Collapse
|
21
|
Zheng DF, Zha XJ, Jiang EL, Qiu Y, Yang W, Xiao WD. Trojan Horse-Like Biohybrid Nanozyme for Ameliorating Liver Ischemia-Reperfusion Injury. Adv Healthc Mater 2025; 14:e2404458. [PMID: 39828639 DOI: 10.1002/adhm.202404458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 01/08/2025] [Indexed: 01/22/2025]
Abstract
Liver ischemia and reperfusion (I/R) injury is a reactive oxygen species (ROS)-related disease that occurs during liver transplantation and resection and hinders postoperative liver function recovery. Current approaches to alleviate liver I/R injury have limited effectiveness due to the short circulation time, poor solubility, and severe side effects of conventional antioxidants and anti-inflammatory drugs. Herein, a universal strategy is proposed to fabricate a Trojan horse-like biohybrid nanozyme (THBN) with hepatic-targeting capabilities. Tannic acid (TA) mediates adeno-associated virus (AAV8) decoration onto 2D Ti3C2 nanosheets, resulting in THBN with a size of 116.2 ± 9.5 nm. Remarkably, THBN exhibits catalase (CAT)-like activity, broad-spectrum ROS scavenging activity and targeted delivery to liver tissue owing to the presence of AAV8. Both in vivo and in vitro experiments confirmed the efficacy of THBN in attenuating liver I/R injury by mitigating inflammation and oxidative stress and inhibiting hepatocellular apoptosis. RNA-seq analysis suggests that THBN may alleviate liver I/R injury by activating the PKC pathway. The effective targeting and therapeutic capabilities of THBN represent an advancement in nanotherapeutics for hepatic ischemia‒reperfusion injury, shedding light on the promising potential of this next-generation nanotherapeutic approach.
Collapse
Affiliation(s)
- Dao-Feng Zheng
- Department of General Surgery, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China
| | - Xiang-Jun Zha
- Department of Ultrasound, Medical Research Center, Affiliated Hospital of Southwest Jiaotong University, The Third People's Hospital of Chengdu, Chengdu, Sichuan, 610031, China
| | - En-Lai Jiang
- Department of General Surgery, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China
| | - Yuan Qiu
- Department of General Surgery, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China
| | - Wei Yang
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, Sichuan, 610065, China
| | - Wei-Dong Xiao
- Department of General Surgery, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China
| |
Collapse
|
22
|
Jiang M, Zhang K, Meng J, Xu L, Liu Y, Wei R. Engineered exosomes in service of tumor immunotherapy: From optimizing tumor-derived exosomes to delivering CRISPR/Cas9 system. Int J Cancer 2025; 156:898-913. [PMID: 39474936 DOI: 10.1002/ijc.35241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 10/09/2024] [Accepted: 10/17/2024] [Indexed: 01/07/2025]
Abstract
Exosomes can be modified and designed for various therapeutic goals because of their unique physical and chemical characteristics. Researchers have identified tumor-derived exosomes (TEXs) as significant players in cancer by influencing tumor growth, immune response evasion, angiogeneis, and drug resistance. TEXs promote the production of specific proteins important for cancer progression. Due to their easy accessibility, TEXs are being modified through genetic, drug delivery, membrane, immune system, and chemical alterations to be repurposed as vehicles for delivering drugs to improve cancer treatment outcomes. In the complex in vivo environment, the clustered regularly interspaced short palindromic repeats and CRISPR-associated protein 9 (CRISPR/Cas9) system encounters challenges from degradation, neutralization, and immune responses, emphasizing the need for strategic distribution strategies for effective genome editing. Engineered exosomes present a promising avenue for delivering CRISPR/Cas9 in vivo. In this review, we will explore different techniques for enhancing TEXs using various engineering strategies. Additionally, we will discuss how these exosomes can be incorporated into advanced genetic engineering systems like CRISPR/Cas9 for possible therapeutic uses.
Collapse
Affiliation(s)
- Mingyang Jiang
- Department of Bone and Joint Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Ke Zhang
- Department of Bone and Joint Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Jinfeng Meng
- The First Clinical Medical College, Guangxi Medical University, Nanning
| | - Linhua Xu
- The First Clinical Medical College, Guangxi Medical University, Nanning
| | - Ying Liu
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Ruqiong Wei
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| |
Collapse
|
23
|
Yang M, Xu L, Wang W. Molecular anti-tumorigenic mechanism of Radix Polygoni Multiflori-derived exosome-like nanoparticles. Heliyon 2025; 11:e41918. [PMID: 40028544 PMCID: PMC11868940 DOI: 10.1016/j.heliyon.2025.e41918] [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: 04/19/2024] [Revised: 11/25/2024] [Accepted: 01/10/2025] [Indexed: 03/05/2025] Open
Abstract
The incidence and mortality rate of liver cancer are on the rise worldwide and effective therapeutic strategies are needed. Previous studies demonstrated that exosomes and exosome-like nanoparticles (ELNs) have numerous biological activities including antioxidant, anti-tumor, and anti-inflammatory effects. In this study, ELNs were successfully isolated for the first time from Radix Polygoni Multiflori (RPM) juice by ultracentrifugation. This indicated that RPM-ELNs could be internalized by hepatoma cells effectively and inhibit their proliferation and migration in vitro. RNA sequencing and qPCR experiments were performed, and the results of bioinformatic analyses revealed that RPM-ELNs could globally regulate expression levels of numerous genes associated with cancer and the cell cycle. In addition, it showed that RPM-ELNs mainly target liver tissue of mice in vivo.
Collapse
Affiliation(s)
- Mingqing Yang
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Lining Xu
- Department of General Surgery, The Second Medical Center & National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing, China
| | - Weiyu Wang
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, National Quality Control Center for Donated Organ Procurement, Hubei Key Laboratory of Medical Technology on Transplantation, Wuhan, Hubei, China
| |
Collapse
|
24
|
Huang C, Hu Q, Wang P, Xie M, Zhang Y, Li Z, Tang S, Zhang Y, Tian Z, Liu X, Hu Z, Liang D. Overexpression of NKG2D and IL24 in NK Cell-Derived Exosomes for Cancer Therapy. Int J Mol Sci 2025; 26:2098. [PMID: 40076725 PMCID: PMC11901126 DOI: 10.3390/ijms26052098] [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: 01/29/2025] [Revised: 02/23/2025] [Accepted: 02/24/2025] [Indexed: 03/14/2025] Open
Abstract
Natural killer (NK) cell-derived exosomes (NK-Exos) are emerging as a promising avenue in cancer immunotherapy due to their inherent tumor-targeting properties and their capacity to deliver therapeutic agents directly to malignant cells. This research delves into the boosted anti-tumor potency of NK-Exos that has been genetically enhanced to overexpress NKG2D, a vital activating receptor, along with interleukin-24 (IL24), a cytokine renowned for its selective suppressive impact on tumor cells. NKG2D facilitates the recognition of tumor cells by binding to stress-induced ligands, while IL24 induces apoptosis and modulates immune responses to enhance tumor destruction. The NK-Exos engineered to express both NKG2D and IL24 significantly enhanced tumor targeting and increased the apoptosis rate of tumor cells by 30% in A549 and by 20% in HELA at 48 h compared with non-modified NK-Exos, respectively. Furthermore, this enhancement also impacted cell proliferation, with inhibition rates increasing by 30%, 15%, and 15% in A549, HELA, and MCF-7 cells, respectively, and it reduced A549 cell migration by 10%. The integration of NKG2D and IL24 within NK-Exos confers a dual therapeutic mechanism, synergistically amplifying their efficacy in cancer treatment. The utility of NK-Exos co-expressing NKG2D and IL24 offers a novel approach to overcome the limitations of current therapies, providing prolonged tumor suppression and precise targeting of malignant cells and holding great promise for clinical application.
Collapse
Affiliation(s)
- Chujun Huang
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha 410078, China; (C.H.); (Q.H.); (P.W.); (M.X.); (Y.Z.); (Z.L.); (S.T.); (Y.Z.); (Z.T.); (X.L.)
| | - Qian Hu
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha 410078, China; (C.H.); (Q.H.); (P.W.); (M.X.); (Y.Z.); (Z.L.); (S.T.); (Y.Z.); (Z.T.); (X.L.)
| | - Peiyun Wang
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha 410078, China; (C.H.); (Q.H.); (P.W.); (M.X.); (Y.Z.); (Z.L.); (S.T.); (Y.Z.); (Z.T.); (X.L.)
| | - Mi Xie
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha 410078, China; (C.H.); (Q.H.); (P.W.); (M.X.); (Y.Z.); (Z.L.); (S.T.); (Y.Z.); (Z.T.); (X.L.)
| | - Ying Zhang
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha 410078, China; (C.H.); (Q.H.); (P.W.); (M.X.); (Y.Z.); (Z.L.); (S.T.); (Y.Z.); (Z.T.); (X.L.)
| | - Zhixing Li
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha 410078, China; (C.H.); (Q.H.); (P.W.); (M.X.); (Y.Z.); (Z.L.); (S.T.); (Y.Z.); (Z.T.); (X.L.)
| | - Shuqing Tang
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha 410078, China; (C.H.); (Q.H.); (P.W.); (M.X.); (Y.Z.); (Z.L.); (S.T.); (Y.Z.); (Z.T.); (X.L.)
| | - Yuxuan Zhang
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha 410078, China; (C.H.); (Q.H.); (P.W.); (M.X.); (Y.Z.); (Z.L.); (S.T.); (Y.Z.); (Z.T.); (X.L.)
| | - Zhixin Tian
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha 410078, China; (C.H.); (Q.H.); (P.W.); (M.X.); (Y.Z.); (Z.L.); (S.T.); (Y.Z.); (Z.T.); (X.L.)
| | - Xionghao Liu
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha 410078, China; (C.H.); (Q.H.); (P.W.); (M.X.); (Y.Z.); (Z.L.); (S.T.); (Y.Z.); (Z.T.); (X.L.)
| | - Zhiqing Hu
- MOE Key Lab of Rare Pediatric Diseases & Department of Cell Biology and Genetics, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang 421001, China
| | - Desheng Liang
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha 410078, China; (C.H.); (Q.H.); (P.W.); (M.X.); (Y.Z.); (Z.L.); (S.T.); (Y.Z.); (Z.T.); (X.L.)
| |
Collapse
|
25
|
Yang S, Zhu H, Jin H, Wang K, Song J, Sun N, Liu Y, Yin X, Wang R, Wu X, Liu H, Zhang C, Zhao W, Yu F. Bio-orthogonal-labeled exosomes reveals specific distribution in vivo and provides potential application in ARDS therapy. Biomaterials 2025; 319:123208. [PMID: 40023928 DOI: 10.1016/j.biomaterials.2025.123208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 02/15/2025] [Accepted: 02/23/2025] [Indexed: 03/04/2025]
Abstract
Exosomes derived from specific cells may be useful for targeted drug delivery, but tracking them in vivo is essential for their clinical application. However, their small size and complex structure challenge the development of exosome-tracking techniques, and traditional labeling methods are limited by weak affinity and potential toxicity. To address these issues, here we developed a novel bio-orthogonal labeling strategy based on phosphatidylinositol derivatives to fluorescently label exosomes from various human and mouse cell types. The different cell-derived exosomes revealed organ-specific distribution patterns and a favorable safety profile. Notably, 4T1 cell-derived exosomes specifically targeted the lungs. When used as drug carriers loaded with anti-inflammatory resveratrol, these exosomes showed significant therapeutic efficacy in mice with acute respiratory distress syndrome (ARDS), effectively reducing inflammatory responses, mitigating pulmonary fibrosis, and restoring lung tissue morphology and function. Our findings provide a novel exosome labeling strategy and an invaluable tool for their in vivo tracking and targeting screening, while exosomes that specifically target the lungs offer a potential therapeutic strategy for organ-specific diseases such as ARDS.
Collapse
Affiliation(s)
- Song Yang
- Qingdao Central Hospital, School of Health and Life Sciences, University of Health and Rehabilitation Sciences, No. 369, Qingdao National High-Tech Industrial Development Zone, Qingdao, 266113, China; State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Key Laboratory of Molecular Drug Research and KLMDASR of Tianjin, Nankai University, No.38 Tongyan Road, Haihe Education Park, Tianjin, 300350, China
| | - Haomiao Zhu
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Key Laboratory of Molecular Drug Research and KLMDASR of Tianjin, Nankai University, No.38 Tongyan Road, Haihe Education Park, Tianjin, 300350, China; Department of Pharmacy, Qilu Hospital, Shandong University, No.107 Cultural West Road, Jinan, 250012, China
| | - Hongzhen Jin
- Qingdao Central Hospital, School of Health and Life Sciences, University of Health and Rehabilitation Sciences, No. 369, Qingdao National High-Tech Industrial Development Zone, Qingdao, 266113, China; State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Key Laboratory of Molecular Drug Research and KLMDASR of Tianjin, Nankai University, No.38 Tongyan Road, Haihe Education Park, Tianjin, 300350, China
| | - Kun Wang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Key Laboratory of Molecular Drug Research and KLMDASR of Tianjin, Nankai University, No.38 Tongyan Road, Haihe Education Park, Tianjin, 300350, China
| | - Junna Song
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Key Laboratory of Molecular Drug Research and KLMDASR of Tianjin, Nankai University, No.38 Tongyan Road, Haihe Education Park, Tianjin, 300350, China
| | - Na Sun
- Qingdao Central Hospital, School of Health and Life Sciences, University of Health and Rehabilitation Sciences, No. 369, Qingdao National High-Tech Industrial Development Zone, Qingdao, 266113, China
| | - Yonghui Liu
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Key Laboratory of Molecular Drug Research and KLMDASR of Tianjin, Nankai University, No.38 Tongyan Road, Haihe Education Park, Tianjin, 300350, China; School of Chemistry, Tiangong University, No.399 BinShuiXi Road, Tianjin, 300387, China
| | - Xiaona Yin
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Key Laboratory of Molecular Drug Research and KLMDASR of Tianjin, Nankai University, No.38 Tongyan Road, Haihe Education Park, Tianjin, 300350, China
| | - Rui Wang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Key Laboratory of Molecular Drug Research and KLMDASR of Tianjin, Nankai University, No.38 Tongyan Road, Haihe Education Park, Tianjin, 300350, China
| | - Xiao Wu
- Qingdao Central Hospital, School of Health and Life Sciences, University of Health and Rehabilitation Sciences, No. 369, Qingdao National High-Tech Industrial Development Zone, Qingdao, 266113, China
| | - Huadong Liu
- Qingdao Central Hospital, School of Health and Life Sciences, University of Health and Rehabilitation Sciences, No. 369, Qingdao National High-Tech Industrial Development Zone, Qingdao, 266113, China
| | - Chunling Zhang
- Qingdao Central Hospital, School of Health and Life Sciences, University of Health and Rehabilitation Sciences, No. 369, Qingdao National High-Tech Industrial Development Zone, Qingdao, 266113, China.
| | - Wei Zhao
- Qingdao Central Hospital, School of Health and Life Sciences, University of Health and Rehabilitation Sciences, No. 369, Qingdao National High-Tech Industrial Development Zone, Qingdao, 266113, China; State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Key Laboratory of Molecular Drug Research and KLMDASR of Tianjin, Nankai University, No.38 Tongyan Road, Haihe Education Park, Tianjin, 300350, China.
| | - Fan Yu
- Qingdao Central Hospital, School of Health and Life Sciences, University of Health and Rehabilitation Sciences, No. 369, Qingdao National High-Tech Industrial Development Zone, Qingdao, 266113, China; State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Key Laboratory of Molecular Drug Research and KLMDASR of Tianjin, Nankai University, No.38 Tongyan Road, Haihe Education Park, Tianjin, 300350, China.
| |
Collapse
|
26
|
Luo X, McAndrews KM, Kalluri R. Natural and Bioengineered Extracellular Vesicles in Diagnosis, Monitoring and Treatment of Cancer. ACS NANO 2025; 19:5871-5896. [PMID: 39869032 DOI: 10.1021/acsnano.4c11630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
Extracellular vesicles (EVs) are cell derived nanovesicles which are implicated in both physiological and pathological intercellular communication, including the initiation, progression, and metastasis of cancer. The exchange of biomolecules between stromal cells and cancer cells via EVs can provide a window to monitor cancer development in real time for better diagnostic and interventional strategies. In addition, the process of secretion and internalization of EVs by stromal and cancer cells in the tumor microenvironment (TME) can be exploited for delivering therapeutics. EVs have the potential to provide a targeted, biocompatible, and efficient delivery platform for the treatment of cancer and other diseases. Natural as well as engineered EVs as nanomedicine have immense potential for disease intervention. Here, we provide an overview of current knowledge of EVs' function in cancer progression, diagnostic and therapeutic applications for EVs in the cancer setting, as well as current EV engineering strategies.
Collapse
Affiliation(s)
- Xin Luo
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, Texas 77054, United States
- Department of Bioengineering, Rice University, Houston, Texas 77005, United States
| | - Kathleen M McAndrews
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, Texas 77054, United States
| | - Raghu Kalluri
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, Texas 77054, United States
- Department of Bioengineering, Rice University, Houston, Texas 77005, United States
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas 77030, United States
| |
Collapse
|
27
|
Cheng L, Luo P, Li W, Chen Q, Gan L, Zhang F, Zheng L, Qian B. Research progress of extracellular vesicles in the pathogenesis of type IIIA chronic prostatitis. Front Immunol 2025; 16:1496055. [PMID: 40034709 PMCID: PMC11873842 DOI: 10.3389/fimmu.2025.1496055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Accepted: 01/27/2025] [Indexed: 03/05/2025] Open
Abstract
Chronic prostatitis is a prevalent urological condition that significantly impacts patients' quality of life. Advances in the study of Extracellular Vesicles (EV) have revealed their close involvement in the pathogenesis of prostatitis. This paper reviews the progress in understanding the role of EV in the pathogenesis of chronic prostatitis type IIIA, particularly their involvement in inflammatory responses, cell signaling, and interactions with immune cells. Additionally, it explores the potential applications of EV as drug delivery vehicles, including the targeted delivery of anti-inflammatory agents and immunomodulators, and highlights the challenges associated with developing exosome-based therapeutic strategies. In-depth research on EV holds promise for offering new insights into the diagnosis and treatment of inflammatory diseases.
Collapse
Affiliation(s)
- Le Cheng
- The First Clinical College, Gannan Medical University, Ganzhou, Jiangxi, China
- Department of Urology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
- Key Laboratory of Urology and Andrology of Ganzhou, Institute of Urology, First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Peiyue Luo
- The First Clinical College, Gannan Medical University, Ganzhou, Jiangxi, China
- Department of Urology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
- Key Laboratory of Urology and Andrology of Ganzhou, Institute of Urology, First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Wei Li
- The First Clinical College, Gannan Medical University, Ganzhou, Jiangxi, China
- Department of Urology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
- Key Laboratory of Urology and Andrology of Ganzhou, Institute of Urology, First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Qi Chen
- The First Clinical College, Gannan Medical University, Ganzhou, Jiangxi, China
- Department of Urology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
- Key Laboratory of Urology and Andrology of Ganzhou, Institute of Urology, First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Lifeng Gan
- The First Clinical College, Gannan Medical University, Ganzhou, Jiangxi, China
- Department of Urology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
- Key Laboratory of Urology and Andrology of Ganzhou, Institute of Urology, First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Fangtao Zhang
- The First Clinical College, Gannan Medical University, Ganzhou, Jiangxi, China
- Department of Urology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
- Key Laboratory of Urology and Andrology of Ganzhou, Institute of Urology, First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Liying Zheng
- Department of Graduate, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Biao Qian
- Department of Urology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
- Key Laboratory of Urology and Andrology of Ganzhou, Institute of Urology, First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| |
Collapse
|
28
|
Ma C, Tang W, Wang J, Yang S, Hou J, Guo M, Hao L. Application of engineered exosomes in tumor therapy. Am J Transl Res 2025; 17:736-747. [PMID: 40092132 PMCID: PMC11909558 DOI: 10.62347/kixf4662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Accepted: 01/09/2025] [Indexed: 03/19/2025]
Abstract
Malignant tumors pose a significant threat to human health, and conventional cancer therapies are limited by inadequate targeting, leading to severe side effects. Exosomes, as extracellular vesicles mediating intercellular communication, exhibit advantages such as low immunogenicity, high biocompatibility, and low toxicity. After modification, engineered exosomes can be employed as targeted delivery vehicles in tumor therapy. This review summarizes the cellular origin, production methods, engineering strategies, and drug-loading routes of engineered exosomes, discusses their applications in cancer treatment, and delves into the challenges and issues in translating engineered exosomes to clinical practice, aiming to provide insights for exosome engineering research.
Collapse
Affiliation(s)
- Chunhui Ma
- Faculty of Medical Imaging, Naval Medical UniversityShanghai 200433, China
| | - Wei Tang
- School of Basic Medicine, Naval Medical UniversityShanghai 200433, China
| | - Jiaye Wang
- School of Basic Medicine, Naval Medical UniversityShanghai 200433, China
| | - Shiyu Yang
- School of Basic Medicine, Naval Medical UniversityShanghai 200433, China
| | - Jin Hou
- National Key Laboratory of Immunity and Inflammation, Institute of Immunology, Naval Medical UniversityShanghai 200433, China
| | - Meng Guo
- National Key Laboratory of Immunity and Inflammation, Institute of Immunology, Naval Medical UniversityShanghai 200433, China
| | - Lu Hao
- Faculty of Medical Imaging, Naval Medical UniversityShanghai 200433, China
| |
Collapse
|
29
|
Morabbi A, Karimian M. Therapeutic potential of exosomal lncRNAs derived from stem cells in wound healing: focusing on mesenchymal stem cells. Stem Cell Res Ther 2025; 16:62. [PMID: 39934913 DOI: 10.1186/s13287-025-04200-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Accepted: 01/30/2025] [Indexed: 02/13/2025] Open
Abstract
The self-renewal ability and multipotency of stem cells give them great potential for use in wound healing. Stem cell-derived exosomes, owing to their close biological resemblance to their parent cells, offer a more efficient, safer, and economical approach for facilitating cellular communication and interactions within different environments. This potential makes them particularly valuable in the treatment of both acute and chronic wounds, such as lacerations, burns, and diabetic ulcers. Long non-coding RNAs (lncRNAs) enclosed in exosomes, as one of the leading actors of these extracellular microvesicles, through interaction with miRNAs and regulation of various signaling pathways involved in inflammation, angiogenesis, cell proliferation, and migration, could heal the wounds. Exosome-derived lncRNAs from stem cells facilitate extracellular matrix remodeling through interaction between macrophages and fibroblasts. Moreover, alongside regulating the expression of inflammatory cytokines, controlling reactive oxygen species levels, and enhancing autophagic activity, they also modulate immune responses to support wound healing. Regulating the expression of genes and signaling pathways related to angiogenesis, by increasing blood supply and accelerating the delivery of essential substances to the wound environment, is another effect exosomal lncRNAs derived from stem cells for wound healing. These lncRNAs can also enhance skin wound healing by regulating homeostasis, increasing the proliferation and differentiation of cells involved in the wound-healing process, and enhancing fibroblast viability and migration to the injury site. Ultimately, exosome-derived lncRNAs from stem cells offer valuable and novel insights into the molecular mechanisms underlying improved wound healing. They can pave the way for potential therapeutic strategies, fostering further research for a better future. Meanwhile, exosomes derived from mesenchymal stem cells, due to their exceptional regenerative properties, as well as the lncRNAs derived from these exosomes, have emerged as one of the innovative tools in wound healing. This review article aims to narrate the cellular and molecular roles of exosome-derived lncRNAs from stem cells in enhancing wound healing with a focus on mesenchymal stem cells.
Collapse
Affiliation(s)
- Ali Morabbi
- Department of Molecular and Cell Biology, Faculty of Basic Sciences, University of Mazandaran, Babolsar, 47416-95447, Iran
| | - Mohammad Karimian
- Department of Molecular and Cell Biology, Faculty of Basic Sciences, University of Mazandaran, Babolsar, 47416-95447, Iran.
| |
Collapse
|
30
|
Chen C, Chen Z, Zhao J, Wen X, Yao H, Weng Z, Xiong H, Zheng Z, Wu J. TMEM45A enhances palbociclib resistance and cellular glycolysis by activating AKT/mTOR signaling pathway in HR+ breast cancer. Cell Death Discov 2025; 11:47. [PMID: 39910045 PMCID: PMC11799145 DOI: 10.1038/s41420-025-02336-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 12/25/2024] [Accepted: 01/28/2025] [Indexed: 02/07/2025] Open
Abstract
Palbociclib, a CDK4/6 inhibitor, plays a crucial role in the treatment of HR+ breast cancer. However, resistance to palbociclib is a significant concern that merits further investigation. Our investigation identifies TMEM45A as a potential driver of palbociclib resistance and its association with increased cellular glycolysis. We demonstrate that TMEM45A is highly expressed in palbociclib-resistant breast cancer (BRCA) cells, correlating with enhanced tumor progression. Silencing TMEM45A enhances sensitivity to palbociclib, promotes cell cycle arrest and apoptosis, and inhibits the proliferation of BRCA cells. Moreover, attenuation of TMEM45A expression reduces cancer aggressiveness by decreasing the expression of EMT and glycolysis-related proteins. Subsequent gene set enrichment analysis (GSEA) confirms that TMEM45A activates the AKT/mTOR signaling pathway, which is integral to cell cycle progression and glycolysis. In a cell line-derived xenograft (CDX) mouse model, TMEM45A knockdown significantly restores sensitivity to palbociclib and suppresses tumor growth. Additionally, the use of engineered exosomes loaded with siRNA targeting TMEM45A presents a promising strategy for enhancing CDK4/6 inhibitor sensitivity without observable toxic side effects in a patient-derived xenograft (PDX) model. Collectively, our findings suggest that TMEM45A may be a therapeutic target for overcoming palbociclib resistance, and exosomal siRNA delivery could be a viable strategy for precision medicine in HR+ breast cancer.
Collapse
Affiliation(s)
- Cui Chen
- Department of Thyroid and Breast Surgery, The Third Affiliated Hospital of Sun Yat-Sen University, Sun Yat-sen University, Guangzhou, China
- Department of Gastrointestinal Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Sun Yat-sen University, Guangzhou, China
| | - Zehong Chen
- Department of Gastrointestinal Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Sun Yat-sen University, Guangzhou, China
| | - Jinze Zhao
- Department of Thyroid and Breast Surgery, The Third Affiliated Hospital of Sun Yat-Sen University, Sun Yat-sen University, Guangzhou, China
| | - Xinyun Wen
- Department of Thyroid and Breast Surgery, The Third Affiliated Hospital of Sun Yat-Sen University, Sun Yat-sen University, Guangzhou, China
| | - Hanming Yao
- Department of Gastroenterology, Guangdong Provincial Geriatrics Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China
| | - Zijin Weng
- Department of Pathology, The Third Affiliated Hospital of Sun Yat-sen University, Sun Yat-sen University, Guangzhou, China
| | - Huiping Xiong
- Department of Thyroid and Breast Surgery, The Third Affiliated Hospital of Sun Yat-Sen University, Sun Yat-sen University, Guangzhou, China
| | - Zongheng Zheng
- Department of Gastrointestinal Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Sun Yat-sen University, Guangzhou, China.
| | - Juekun Wu
- Department of Thyroid and Breast Surgery, The Third Affiliated Hospital of Sun Yat-Sen University, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
31
|
Sun X, Chen X, Ni Y, Li X, Song J, Wang J, Xu S, Shu W, Chen M. Latexin (LXN) enhances tumor immune surveillance in mice by inhibiting Treg cells through the macrophage exosome pathway. Int J Biol Macromol 2025; 289:138822. [PMID: 39694381 DOI: 10.1016/j.ijbiomac.2024.138822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 12/12/2024] [Accepted: 12/14/2024] [Indexed: 12/20/2024]
Abstract
Latexin (LXN) is a secreted protein with a molecular weight of 29 KD, which is considered a tumor suppressor and plays an important role in the inflammatory immune response. LXN is highly expressed in macrophages and regulates macrophage polarity and tumor immune escape, demonstrating excellent clinical potential. However, its mechanism is still unclear. In this study, a macrophage-T cell co-culture system is established to clarify the secretion of macrophage LXN into the extracellular through exosomes. The results indicate that LXN in macrophage-derived exosomes is functional, that is, LXN-enriched exosome inhibits CD4+T cell differentiation into Treg cells in vitro and in vivo, and exhibits good tumor suppressive effects. Based on this discovery, a biomimetic nanoparticle loaded with LXN protein (MØ@LXN-NPS) is designed and synthesized. Furthermore, the MØ@LXN-NPS shows excellent performance in both in vivo and in vitro, especially in enhancing tumor immune surveillance by inhibiting Treg cells in tumor microenvironment, thus exhibiting excellent anti-tumor activity. This study provides a demonstration for the transition of biomolecules from functional research to engineering applications. The excellent performance of MØ@LXN-NPS in tumor immune regulation suggests that the engineered biomimetic nanomedicine has good clinical application prospects.
Collapse
Affiliation(s)
- Xuchen Sun
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, China
| | - Xuanming Chen
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, China
| | - Yuanting Ni
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, China
| | - Xiuzhen Li
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, China
| | - Jiaqi Song
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, China
| | - Jingzhu Wang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, China
| | - Shaohua Xu
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, China
| | - Wei Shu
- College of Intelligent Medicine and Biotechnology, Guilin Medical University, Guilin, China
| | - Ming Chen
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, China.
| |
Collapse
|
32
|
Li X, Zhao H. Targeting secretory autophagy in solid cancers: mechanisms, immune regulation and clinical insights. Exp Hematol Oncol 2025; 14:12. [PMID: 39893499 PMCID: PMC11786567 DOI: 10.1186/s40164-025-00603-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Accepted: 01/25/2025] [Indexed: 02/04/2025] Open
Abstract
Secretory autophagy is a classical form of unconventional secretion that integrates autophagy with the secretory process, relying on highly conserved autophagy-related molecules and playing a critical role in tumor progression and treatment resistance. Traditional autophagy is responsible for degrading intracellular substances by fusing autophagosomes with lysosomes. However, secretory autophagy uses autophagy signaling to mediate the secretion of specific substances and regulate the tumor microenvironment (TME). Cytoplasmic substances are preferentially secreted rather than directed toward lysosomal degradation, involving various selective mechanisms. Moreover, substances released by secretory autophagy convey biological signals to the TME, inducing immune dysregulation and contributing to drug resistance. Therefore, elucidating the mechanisms underlying secretory autophagy is essential for improving clinical treatments. This review systematically summarizes current knowledge of secretory autophagy, from initiation to secretion, considering inter-tumor heterogeneity, explores its role across different tumor types. Furthermore, it proposes future research directions and highlights unresolved clinical challenges.
Collapse
Affiliation(s)
- Xinyu Li
- Department of General Surgery, Fourth Affiliated Hospital of China Medical University, Shenyang City, 110032, Liaoning Province, China
| | - Haiying Zhao
- Department of General Surgery, Fourth Affiliated Hospital of China Medical University, Shenyang City, 110032, Liaoning Province, China.
| |
Collapse
|
33
|
Huang J, Li H, Mei Y, Yi P, Ren Y, Wang Y, Han L, Tang Q, Liu D, Chen W, An Y, Hu C. An Injectable Hydrogel Bioimplant Loaded with Engineered Exosomes and Triple Anti-Tuberculosis Drugs with Potential for Treating Bone and Joint Tuberculosis. Int J Nanomedicine 2025; 20:1285-1302. [PMID: 39911262 PMCID: PMC11794387 DOI: 10.2147/ijn.s480288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 12/12/2024] [Indexed: 02/07/2025] Open
Abstract
Purpose Treatment for bone and joint tuberculosis (BJTB) is challenging due to its refractory and recurrent nature. This study aimed to develop a bioimplantable scaffold with osteoinductive and antituberculosis characteristics to treat BJTB. Methods This scaffold is built on oxidized hyaluronic acid and carboxymethyl chitosan hydrogel mixed with hydroxyapatite as a bone tissue engineered material. In order to make the scaffold have the biological activity of promoting tissue repair, the engineered exosomes (Exoeng) were added innovatively. In addition, drug-loaded liposomes equipped with an aldehyde group on the surface are cross-linked with the amine group of the hydrogel skeleton to participate in the Schiff base reaction. Results The designed scaffold has characteristics of self-healing and injectability exhibit excellent anti-tuberculosis and promoting bone repair activities. Exoeng strongly stimulates cellular angiogenesis and osteogenic differentiation. The liposomes coated in hydrogel can release three kinds of anti-tuberculosis drugs smoothly and slowly, achieving a long term anti-tuberculosis. Conclusion The composite bio-scaffold shows good tissue repair and long-term anti-tuberculosis abilities, which expected to provide a viable treatment plan for bone-related BJTB.
Collapse
Affiliation(s)
- Jiayan Huang
- Department of Tuberculosis, the Second Hospital of Nanjing, Affiliated to Nanjing University of Chinese Medicine, Nanjing, People’s Republic of China
| | - Han Li
- Department of Pharmacy, Central Hospital of Guangdong Provincial Nongken, Zhanjiang, Guangdong, People’s Republic of China
| | - Yuting Mei
- Department of Tuberculosis, the Second Hospital of Nanjing, Affiliated to Nanjing University of Chinese Medicine, Nanjing, People’s Republic of China
| | - Pengcheng Yi
- Department of Tuberculosis, the Second Hospital of Nanjing, Affiliated to Nanjing University of Chinese Medicine, Nanjing, People’s Republic of China
| | - Yunyao Ren
- Department of Tuberculosis, the Second Hospital of Nanjing, Affiliated to Nanjing University of Chinese Medicine, Nanjing, People’s Republic of China
| | - Yunjuan Wang
- Medical School of Southeast University, Nanjing, People’s Republic of China
| | - Limei Han
- Department of Tuberculosis, the Second Hospital of Nanjing, Affiliated to Nanjing University of Chinese Medicine, Nanjing, People’s Republic of China
| | - Qiusha Tang
- Medical School of Southeast University, Nanjing, People’s Republic of China
| | - Dongfang Liu
- Medical School of Southeast University, Nanjing, People’s Republic of China
| | - Wei Chen
- Department of Clinical Research Center, The Second Hospital of Nanjing, Nanjing University of Chinese Medicine, Nanjing, People’s Republic of China
| | - Yanli An
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology (Southeast University), Center of Interventional Radiology & Vascular Surgery, Department of Radiology, Medical School, Zhong da Hospital, Southeast University, Nanjing, JiangsuPeople’s Republic of China
| | - Chunmei Hu
- Department of Tuberculosis, the Second Hospital of Nanjing, Affiliated to Nanjing University of Chinese Medicine, Nanjing, People’s Republic of China
| |
Collapse
|
34
|
Kareem RA, Sameer HN, Yaseen A, Athab ZH, Adil M, Ahmed HH. A review of the immunomodulatory properties of mesenchymal stem cells and their derived extracellular vesicles in small-cell and non-small-cell lung cancer cells. Int Immunopharmacol 2025; 146:113848. [PMID: 39689606 DOI: 10.1016/j.intimp.2024.113848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 12/09/2024] [Accepted: 12/10/2024] [Indexed: 12/19/2024]
Abstract
Among the most challenging diseases to treat is lung cancer (LC). While immunotherapy has a checkered history, it has lately shown great promise in the treatment of LC, and interest in this promising new approach is on the rise around the globe. Immunotherapy using mesenchymal stem cells (MSCs) is gaining popularity. Regenerative medicine, cell therapy, and immune modulation are three areas that have shown significant interest in MSCs. More than that, MSCs have recently attracted attention as potential anti-cancer drug delivery vehicles due to their inherent ability to go home to tumor locations. Making MSCs a double-edged sword in the fight against neoplastic illnesses, they are also known to impart pro-oncogenic properties. Additionally, multiple studies have proposed extracellular vesicles (EVs) secreted by MSCs as a potential therapeutic agent or method for delivering anti-cancer drugs. However, there has been conflicting evidence regarding the impact of MSCs or MSC-EV on the behavior of cancer cells, and the exact mechanism for this effect is still unknown. Our research has focused on MSCs and their key characteristics, such as their immunomodulatory capabilities for cancer therapy. Our research has also explored the potential of MSCs and their derivatives to treat small-cell and non-small-cell lung cancers (NSCLC and SCLC, respectively) by leveraging MSCs' immunomodulatory characteristics. At the end of this article, we covered the pros and cons of this therapy procedure, as well as what researchers want to do in the future to make it more suitable for clinical application in LC treatment.
Collapse
Affiliation(s)
| | - Hayder Naji Sameer
- Collage of Pharmacy, National University of Science and Technology, Dhi Qar 64001, Iraq
| | | | - Zainab H Athab
- Department of Pharmacy, Al-Zahrawi University College, Karbala, Iraq
| | | | | |
Collapse
|
35
|
Bavafa A, Izadpanahi M, Hosseini E, Hajinejad M, Abedi M, Forouzanfar F, Sahab-Negah S. Exosome: an overview on enhanced biogenesis by small molecules. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-024-03762-9. [PMID: 39862264 DOI: 10.1007/s00210-024-03762-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Accepted: 12/23/2024] [Indexed: 01/27/2025]
Abstract
Exosomes are extracellular vesicles that received attention for their potential use in the treatment of various injuries. They communicate intercellularly by transferring genetic and bioactive molecules from parent cells. Although exosomes hold immense promise for treating neurodegenerative and oncological diseases, their actual clinical use is very limited because of their biogenesis and secretion. Recent studies have shown that small molecules can significantly enhance exosome biogenesis, thereby remarkably improving yield, functionality, and therapeutic effects. These molecules modulate critical pathways toward optimum exosome production in a mode that is either ESCRT dependent or ESCRT independent. Improved exosome biogenesis may provide new avenues for targeted cancer therapy, neuroprotection in neurodegenerative diseases, and regenerative medicine in wound healing. This review explores the role of small molecules in enhancing exosome biogenesis and secretion, highlights their underlying mechanisms, and discusses emerging clinical applications. By addressing current challenges and focusing on translational opportunities, this study provides a foundation for advancing cell-free therapies in regenerative medicine and beyond.
Collapse
Affiliation(s)
- Amir Bavafa
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Neuroscience, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Maryam Izadpanahi
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Neuroscience, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Elham Hosseini
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Neuroscience, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mehrdad Hajinejad
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Qaen Faculty of Medical Sciences, Birjand University of Medical Sciences, Birjand, Iran
| | - Mahsa Abedi
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Institute for Molecular and Clinical Immunology, Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany
| | - Fatemeh Forouzanfar
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
- Department of Neuroscience, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Sajad Sahab-Negah
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
- Department of Neuroscience, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
- Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran, Iran.
| |
Collapse
|
36
|
Lee H, Rho WY, Kim YH, Chang H, Jun BH. CRISPR-Cas9 Gene Therapy: Non-Viral Delivery and Stimuli-Responsive Nanoformulations. Molecules 2025; 30:542. [PMID: 39942646 PMCID: PMC11820414 DOI: 10.3390/molecules30030542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 01/07/2025] [Accepted: 01/17/2025] [Indexed: 02/16/2025] Open
Abstract
The CRISPR-Cas9 technology, one of the groundbreaking genome editing methods for addressing genetic disorders, has emerged as a powerful, precise, and efficient tool. However, its clinical translation remains hindered by challenges in delivery efficiency and targeting specificity. This review provides a comprehensive analysis of the structural features, advantages, and potential applications of various non-viral and stimuli-responsive systems, examining recent progress to emphasize the potential to address these limitations and advance CRISPR-Cas9 therapeutics. We describe how recent reports emphasize that nonviral vectors, including lipid-based nanoparticles, extracellular vesicles, polymeric nanoparticles, gold nanoparticles, and mesoporous silica nanoparticles, can offer diverse advantages to enhance stability, cellular uptake, and biocompatibility, based on their structures and physio-chemical stability. We also summarize recent progress on stimuli-responsive nanoformulations, a type of non-viral vector, to introduce precision and control in CRISPR-Cas9 delivery. Stimuli-responsive nanoformulations are designed to respond to pH, redox states, and external triggers, facilitate controlled and targeted delivery, and minimize off-target effects. The insights in our review suggest future challenges for clinical applications of gene therapy technologies and highlight the potential of delivery systems to enhance CRISPR-Cas9's clinical efficacy, positioning them as pivotal tools for future gene-editing therapies.
Collapse
Affiliation(s)
- Hyunwoo Lee
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 05029, Republic of Korea; (H.L.); (Y.-H.K.)
| | - Won-Yeop Rho
- School of International Engineering and Science, Jeonbuk National University, Jeonju 54896, Republic of Korea;
| | - Yoon-Hee Kim
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 05029, Republic of Korea; (H.L.); (Y.-H.K.)
| | - Hyejin Chang
- Division of Science Education, Kangwon National University, 1 Gangwondaehakgil, Chuncheon-si 24341, Republic of Korea
| | - Bong-Hyun Jun
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 05029, Republic of Korea; (H.L.); (Y.-H.K.)
| |
Collapse
|
37
|
Miao Q, Li S, Lyu W, Zhang J, Han Y. Exosomes in Oral Diseases: Mechanisms and Therapeutic Applications. Drug Des Devel Ther 2025; 19:457-469. [PMID: 39867866 PMCID: PMC11766710 DOI: 10.2147/dddt.s505355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Accepted: 01/06/2025] [Indexed: 01/28/2025] Open
Abstract
Exosomes, small extracellular vesicles secreted by various cells, play crucial roles in the pathogenesis and treatment of oral diseases. Recent studies have highlighted their involvement in orthodontics, periodontitis, oral squamous cell carcinoma (OSCC), and hand, foot, and mouth disease (HFMD). Exosomes have a positive effect on the inflammatory environment of the oral cavity, remodeling and regeneration of oral tissues, and offer promising therapeutic options for bone and periodontal tissue restoration. In OSCC tumor-derived exosomes promote cancer progression through cell proliferation, migration, invasion, and angiogenesis, and serve as potential biomarkers for early diagnosis and prognosis. Additionally, engineered exosomes constructed specifically based on exosome properties hold great promise for targeted drug delivery and regenerative therapies such as bone regeneration in orthodontics and periodontal healing. With continued research, exosomes hold great potential for improving diagnosis and treatment in oral diseases, advancing personalized and regenerative therapies.
Collapse
Affiliation(s)
- Qiandai Miao
- Department of Stomatology, China Academy of Chinese Medical Sciences, Xiyuan Hospital, Beijing, 100091, People’s Republic of China
| | - Shaoqing Li
- Department of Stomatology, China Academy of Chinese Medical Sciences, Xiyuan Hospital, Beijing, 100091, People’s Republic of China
| | - Weijia Lyu
- Department of Stomatology, China Academy of Chinese Medical Sciences, Xiyuan Hospital, Beijing, 100091, People’s Republic of China
| | - Jianxia Zhang
- Department of Stomatology, China Academy of Chinese Medical Sciences, Xiyuan Hospital, Beijing, 100091, People’s Republic of China
| | - Yan Han
- Department of Stomatology, China Academy of Chinese Medical Sciences, Xiyuan Hospital, Beijing, 100091, People’s Republic of China
| |
Collapse
|
38
|
Lin F, Luo H, Wang J, Li Q, Zha L. Macrophage-derived extracellular vesicles as new players in chronic non-communicable diseases. Front Immunol 2025; 15:1479330. [PMID: 39896803 PMCID: PMC11782043 DOI: 10.3389/fimmu.2024.1479330] [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: 08/13/2024] [Accepted: 12/23/2024] [Indexed: 02/04/2025] Open
Abstract
Macrophages are innate immune cells present in all tissues and play an important role in almost all aspects of the biology of living organisms. Extracellular vesicles (EVs) are released by cells and transport their contents (micro RNAs, mRNA, proteins, and long noncoding RNAs) to nearby or distant cells for cell-to-cell communication. Numerous studies have shown that macrophage-derived extracellular vesicles (M-EVs) and their contents play an important role in a variety of diseases and show great potential as biomarkers, therapeutics, and drug delivery vehicles for diseases. This article reviews the biological functions and mechanisms of M-EVs and their contents in chronic non-communicable diseases such as cardiovascular diseases, metabolic diseases, cancer, inflammatory diseases and bone-related diseases. In addition, the potential application of M-EVs as drug delivery systems for various diseases have been summarized.
Collapse
Affiliation(s)
- Fengjuan Lin
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, National Medical Products Administration (NMPA) Key Laboratory for Safety Evaluation of Cosmetics, School of Public Health, Southern Medical University, Guangzhou, Guangdong, China
| | - Huiyu Luo
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, National Medical Products Administration (NMPA) Key Laboratory for Safety Evaluation of Cosmetics, School of Public Health, Southern Medical University, Guangzhou, Guangdong, China
| | - Jiexian Wang
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, National Medical Products Administration (NMPA) Key Laboratory for Safety Evaluation of Cosmetics, School of Public Health, Southern Medical University, Guangzhou, Guangdong, China
| | - Qing Li
- Department of Clinical Nutrition, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Longying Zha
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, National Medical Products Administration (NMPA) Key Laboratory for Safety Evaluation of Cosmetics, School of Public Health, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
39
|
Zhao B, Li Z, Li R. Exosomes in oral squamous cell carcinoma: functions, challenges, and potential applications. Front Oncol 2025; 14:1502283. [PMID: 39886659 PMCID: PMC11779712 DOI: 10.3389/fonc.2024.1502283] [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: 09/26/2024] [Accepted: 12/05/2024] [Indexed: 02/01/2025] Open
Abstract
Oral squamous cell carcinoma (OSCC) accounts for approximately 90% of all oral cancers, significantly impacting the survival and quality of life of patients. Exosomes, small extracellular vesicles released by cells, play a crucial role in intercellular communication in cancer. Nevertheless, their function and mechanism in OSCC remain elusive. Search Pubmed, Web of Science, and Cochrane Library using keywords OSCC, exome, diagnosis, and treatment to review the research progress of exome in OSCC. Based on these results, this review starting from the biosynthesis, structure, and contents of exosomes, elaborates on the research progress of exosomes in the diagnosis and treatment of OSCC. It explores the potential of exosomes in the diagnosis and treatment of OSCC, and briefly describes the challenges researchers currently face.
Collapse
Affiliation(s)
- Bo Zhao
- Key Laboratory of Advanced Intelligent Protective Equipment Technology (Hebei University of Technology), Ministry of Education, Tianjin, China
- Department of Stomatology, Tianjin First Central Hospital, Tianjin, China
| | - Zuntai Li
- Department of Stomatology, Tianjin First Central Hospital, Tianjin, China
| | - Ronghua Li
- Department of Stomatology, Tianjin First Central Hospital, Tianjin, China
| |
Collapse
|
40
|
Zhu S, Shou X, Kuang G, Kong X, Sun W, Zhang Q, Xia J. Stimuli-responsive hydrogel microspheres encapsulated with tumor-cell-derived microparticles for malignant ascites treatment. Acta Biomater 2025; 192:328-339. [PMID: 39586349 DOI: 10.1016/j.actbio.2024.11.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Revised: 11/04/2024] [Accepted: 11/21/2024] [Indexed: 11/27/2024]
Abstract
Tumor-cell-derived microparticles (TMPs) have been recognized as chemotherapeutic drug carriers and immunomodulators for anti-tumor therapy. Research in the clinical application of TMPs has been devoted to developing an effective delivery formulation that could enhance their therapeutic effects. Here, we propose thermal-responsive agarose hydrogel microspheres (MTX-TMPs@MSs) with encapsulation of Methotrexate (MTX)-packaging TMPs (MTX-TMPs) and black phosphorus quantum dots (BPQDs) by microfluidic technology for synergistic treatment of malignant ascites. The laden MTX-TMPs, separated from apoptotic tumor cells, could target tumor cells for the delivery of chemotherapy drugs and modulate the tumor immune microenvironment. Under near-infrared (NIR) induced thermal stimulation, MTX-TMPs could be controllably released from the low-melting-point agarose matrix hydrogel microspheres for chemotherapy (CHT) and immunotherapy (IMT). In addition, benefiting from photothermal therapy (PTT)-induced tumor immunogenic death, the anti-tumor immune response triggered by MTX-TMPs was further enhanced. Based on these features, the MTX-TMPs@MSs could remarkably eliminate tumor cells in vitro and obviously suppress tumor growth in vivo through synergistic PTT, CHT, and IMT. Therefore, it is envisaged that this TMPs-integrated microcarrier will have promising applications in clinical tumor therapy. STATEMENT OF SIGNIFICANCE: Primary liver cancer ranks third among the causes of cancer deaths globally, with hepatocellular carcinoma (HCC) being the most common type. In particular, patients with advanced HCC accompanied by malignant ascites, a common complication, indicate tumor metastasis and a poor prognosis. In this paper, we developed stimuli-responsive hydrogel microspheres from microfluidics for the delivery of methotrexate (MTX)-loaded tumor-cell-derived microparticles (MTX-TMPs) for synergistic chemotherapy, photothermal therapy, and immunotherapy. The release of MTX-TMPs from hydrogel microspheres could be on-demand controlled through BPQDs-mediated photothermal stimulus. On the other hand, BPQDs-mediated mild hyperthermia cooperatesss with MTX-TMPs-induced chemotherapy could participate in remodeling the tumor immunosuppressive microenvironment. Thus, the prepared microcarrier system holds great promise for tumor therapy.
Collapse
Affiliation(s)
- Shishi Zhu
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China
| | - Xin Shou
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325001, China
| | - Gaizhen Kuang
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China
| | - Xiuyan Kong
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China
| | - Weijian Sun
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China.
| | - Qingfei Zhang
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China; Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325001, China.
| | - Jinglin Xia
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China; National Medical Center & National Clinical Research Center for Interventional Medicine. Liver Cancer Institute, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China.
| |
Collapse
|
41
|
Kim G, Zhu R, Yu S, Fan B, Jeon H, Leon J, Webber MJ, Wang Y. Enhancing Gene Delivery to Breast Cancer with Highly Efficient siRNA Loading and pH-Responsive Small Extracellular Vesicles. ACS Biomater Sci Eng 2025; 11:213-227. [PMID: 39713992 DOI: 10.1021/acsbiomaterials.4c01595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
Small extracellular vesicles (sEVs) are promising nanocarriers for drug delivery to treat a wide range of diseases due to their natural origin and innate homing properties. However, suboptimal therapeutic effects, attributed to ineffective targeting, limited lysosomal escape, and insufficient delivery, remain challenges in effectively delivering therapeutic cargo. Despite advances in sEV-based drug delivery systems, conventional approaches need improvement to address low drug-loading efficiency and to develop surface functionalization techniques for precise targeting of cells of interest, all while preserving the membrane integrity of sEVs. We report an enhanced gene delivery system using multifunctional sEVs for highly efficient siRNA loading and delivery. The integration of chiral graphene quantum dots enhanced the loading capacity while preserving the structural integrity of the sEVs. Additionally, lysosomal escape is facilitated by functionalizing sEVs with pH-responsive peptides, fully harnessing the inherent homing effect of sEVs for targeted and precise delivery. These sEVs achieved a 1.74-fold increase in cytosolic cargo delivery compared to unmodified sEVs, resulting in substantial gene silencing of around 73%. Our approach has significant potential to advance sEV-based gene delivery in order to accelerate clinical progress.
Collapse
Affiliation(s)
- Gaeun Kim
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, Indiana46556, United States
| | - Runyao Zhu
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, Indiana46556, United States
| | - Sihan Yu
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, Indiana46556, United States
| | - Bowen Fan
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, Indiana46556, United States
| | - Hyunsu Jeon
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, Indiana46556, United States
| | - Jennifer Leon
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, Indiana46556, United States
| | - Matthew J Webber
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, Indiana46556, United States
| | - Yichun Wang
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, Indiana46556, United States
| |
Collapse
|
42
|
Huang X, Tang Y. Unveiling the complex double-edged sword role of exosomes in nasopharyngeal carcinoma. PeerJ 2025; 13:e18783. [PMID: 39822977 PMCID: PMC11737332 DOI: 10.7717/peerj.18783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Accepted: 12/09/2024] [Indexed: 01/19/2025] Open
Abstract
Nasopharyngeal carcinoma (NPC) is a malignancy arising from the epithelium of the nasopharynx. Given its late diagnosis, NPC raises serious considerations in Southeast Asia. In addition to resistance to conventional treatment that combines chemotherapy and radiation, NPC has high rates of metastasis and frequent recurrence. Exosomes are small membrane vesicles at the nanoscale that transport physiologically active compounds from their source cell and have a crucial function in signal transmission and intercellular message exchange. The exosomes detected in the tissues of NPC patients have recently emerged as a potential non-invasive liquid biopsy biomarker that plays a role in controlling the tumor pathophysiology. Here, we take a look back at what we know so far about the complex double-edged sword role of exosomes in NPC. Exosomes could serve as biomarkers and therapeutic agents, as well as the molecular mechanisms by which they promote cell growth, angiogenesis, metastasis, immunosuppression, radiation resistance, and chemotherapy resistance in NPC. Furthermore, we go over some of the difficulties and restrictions associated with exosome use. It is anticipated that this article would provide the reference for the apply of exosomes in clinical practice.
Collapse
Affiliation(s)
- Xueyan Huang
- Department of Otorhinolaryngology Head and Neck Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Yuedi Tang
- Department of Otorhinolaryngology Head and Neck Surgery, West China Hospital of Sichuan University, Chengdu, China
| |
Collapse
|
43
|
Peng X, Wang T, Dai B, Zhu Y, Ji M, Yang P, Zhang J, Liu W, Miao Y, Liu Y, Wang S, Sun J. Gene Therapy for Inflammatory Cascade in Intrauterine Injury with Engineered Extracellular Vesicles Hybrid Snail Mucus-enhanced Adhesive Hydrogels. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2410769. [PMID: 39454114 PMCID: PMC11714243 DOI: 10.1002/advs.202410769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Indexed: 10/27/2024]
Abstract
Early hyper-inflammation caused by intrauterine injury triggered subsequent intrauterine adhesion (IUA). STAT1-mediated M1 macrophages are confirmed to secrete pro-inflammatory cytokines to accelerate inflammatory cascade and IUA formation by multi-omics analysis and experimental verification. However, clinically used hyaluronic acid (HA) hydrogels are prone to slip out of injury sites due to poor bio-adhesion properties. Therefore, there are still challenges in applying hydrogels for M1 macrophage intervention in IUA treatment. Herein, an engineered extracellular vesicles (EVs) hybrid snail mucus (SM)-enhanced adhesive hydrogels to improve bio-adhesion property is fabricated and M1 macrophage intervention through targeting delivery and STAT1 silencing is achieved. First, inspired by the high bio-adhesion capacity of SM, SM and gelatin methacrylate (GelMA) solution are mixed to construct GelMA/SM (GS) hydrogel. Then, folic acid-modified extracellular vesicles (FA-EVs) are synthesized for targeting the delivery of STAT1-siRNA. Upon injection of FA-EVs hybrid GS hydrogel into the uterine cavity, a protective hydrogel layer forms on the surface of injury sites and sustains the release of STAT1-siRNA-loaded FA-EVs to curtail M1 macrophages generation through inhibiting STAT1 phosphorylation, resulting in reduction of myofibroblasts activation and collagen deposition. In addition, the pregnancy rate and the number of fetuses in rats treated with this hydrogel were much higher than those in other groups, suggesting that the hydrogel could promote functional endometrial regeneration and restore fertility. Overall, this study presents a promising strategy for employing FA-EVs hybrid adhesive hydrogel with superior bio-adhesion properties and M1 macrophage targeting delivery for IUA treatment and uterus recovery.
Collapse
Affiliation(s)
- Xiaotong Peng
- Department of GynecologyShanghai Key Laboratory of Maternal Fetal MedicineShanghai Institute of Maternal‐Fetal Medicine and Gynecologic OncologyShanghai First Maternity and Infant HospitalSchool of MedicineTongji UniversityShanghai200092China
| | - Tao Wang
- Department of GynecologyShanghai Key Laboratory of Maternal Fetal MedicineShanghai Institute of Maternal‐Fetal Medicine and Gynecologic OncologyShanghai First Maternity and Infant HospitalSchool of MedicineTongji UniversityShanghai200092China
| | - Bo Dai
- Department of HematologyHuashan HospitalFudan UniversityShanghai200040China
| | - Yiping Zhu
- Department of GynecologyShanghai Key Laboratory of Maternal Fetal MedicineShanghai Institute of Maternal‐Fetal Medicine and Gynecologic OncologyShanghai First Maternity and Infant HospitalSchool of MedicineTongji UniversityShanghai200092China
| | - Mei Ji
- Department of GynecologyShanghai Key Laboratory of Maternal Fetal MedicineShanghai Institute of Maternal‐Fetal Medicine and Gynecologic OncologyShanghai First Maternity and Infant HospitalSchool of MedicineTongji UniversityShanghai200092China
| | - Pusheng Yang
- Department of GynecologyShanghai Key Laboratory of Maternal Fetal MedicineShanghai Institute of Maternal‐Fetal Medicine and Gynecologic OncologyShanghai First Maternity and Infant HospitalSchool of MedicineTongji UniversityShanghai200092China
| | - Jiaxin Zhang
- Department of GynecologyShanghai Key Laboratory of Maternal Fetal MedicineShanghai Institute of Maternal‐Fetal Medicine and Gynecologic OncologyShanghai First Maternity and Infant HospitalSchool of MedicineTongji UniversityShanghai200092China
| | - Wenwen Liu
- Department of GynecologyShanghai Key Laboratory of Maternal Fetal MedicineShanghai Institute of Maternal‐Fetal Medicine and Gynecologic OncologyShanghai First Maternity and Infant HospitalSchool of MedicineTongji UniversityShanghai200092China
| | - Yaxin Miao
- Department of GynecologyShanghai Key Laboratory of Maternal Fetal MedicineShanghai Institute of Maternal‐Fetal Medicine and Gynecologic OncologyShanghai First Maternity and Infant HospitalSchool of MedicineTongji UniversityShanghai200092China
| | - Yonghang Liu
- School of Pharmacy and State Key Laboratory of Quality Research in Chinese MedicineMacau University of Science and TechnologyMacao999078China
| | - Shuo Wang
- Department of OrthopaedicsShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghai200233China
| | - Jing Sun
- Department of GynecologyShanghai Key Laboratory of Maternal Fetal MedicineShanghai Institute of Maternal‐Fetal Medicine and Gynecologic OncologyShanghai First Maternity and Infant HospitalSchool of MedicineTongji UniversityShanghai200092China
| |
Collapse
|
44
|
Wang L, Liu H, Chen G, Wu Q, Xu S, Zhou Q, Zhao Y, Wang Q, Yan T, Cheng X. Bubble Ticket Trip: Exploring the Mechanism of miRNA Sorting into Exosomes and Maintaining the Stability of Tumor Microenvironment. Int J Nanomedicine 2024; 19:13671-13685. [PMID: 39723172 PMCID: PMC11669276 DOI: 10.2147/ijn.s498599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Accepted: 12/14/2024] [Indexed: 12/28/2024] Open
Abstract
Exosomes are vesicles ranging from 30 to 100 nanometers in size that show great potential as carriers for therapeutic uses and drug delivery. Enriching a specific set of miRNAs in exosomes emphasizes the existence of particular sorting mechanisms that manage the targeted cargo packaging. The molecular mechanism for miRNA sorting has not been understood. It is crucial to understand the mechanism of exosome encapsulation to develop its therapeutic potential. In this review, we will explore the particular processes through which exosomes naturally encapsulate miRNA, as well as discuss the effect on tumors after encapsulation of miRNAs. We also summarize the effects of targeted drug delivery using genetic engineering and chemical methods to modify exosome-encapsulated miRNA. Finally, gaining insight into how exosome cargo is sorted could be applied in clinical settings for precise drug delivery and to hinder the progression of diseases.
Collapse
Affiliation(s)
- Lu Wang
- Key Laboratory of Cellular Physiology of the Ministry of Education, & Department of Pathology, Shanxi Medical University, Taiyuan, Shanxi Province, 030001, People's Republic of China
| | - Huijuan Liu
- Key Laboratory of Cellular Physiology of the Ministry of Education, & Department of Pathology, Shanxi Medical University, Taiyuan, Shanxi Province, 030001, People's Republic of China
| | - Guohui Chen
- Key Laboratory of Cellular Physiology of the Ministry of Education, & Department of Pathology, Shanxi Medical University, Taiyuan, Shanxi Province, 030001, People's Republic of China
| | - Qinglu Wu
- Key Laboratory of Cellular Physiology of the Ministry of Education, & Department of Pathology, Shanxi Medical University, Taiyuan, Shanxi Province, 030001, People's Republic of China
| | - Songrui Xu
- Key Laboratory of Cellular Physiology of the Ministry of Education, & Department of Pathology, Shanxi Medical University, Taiyuan, Shanxi Province, 030001, People's Republic of China
| | - Qichao Zhou
- Key Laboratory of Cellular Physiology of the Ministry of Education, & Department of Pathology, Shanxi Medical University, Taiyuan, Shanxi Province, 030001, People's Republic of China
| | - Yadong Zhao
- Key Laboratory of Cellular Physiology of the Ministry of Education, & Department of Pathology, Shanxi Medical University, Taiyuan, Shanxi Province, 030001, People's Republic of China
| | - Qiaorong Wang
- Key Laboratory of Cellular Physiology of the Ministry of Education, & Department of Pathology, Shanxi Medical University, Taiyuan, Shanxi Province, 030001, People's Republic of China
| | - Ting Yan
- Key Laboratory of Cellular Physiology of the Ministry of Education, & Department of Pathology, Shanxi Medical University, Taiyuan, Shanxi Province, 030001, People's Republic of China
| | - Xiaolong Cheng
- Key Laboratory of Cellular Physiology of the Ministry of Education, & Department of Pathology, Shanxi Medical University, Taiyuan, Shanxi Province, 030001, People's Republic of China
| |
Collapse
|
45
|
Balaraman AK, Babu MA, Moglad E, Mandaliya V, Rekha MM, Gupta S, Prasad GVS, Kumari M, Chauhan AS, Ali H, Goyal K. Exosome-mediated delivery of CRISPR-Cas9: A revolutionary approach to cancer gene editing. Pathol Res Pract 2024; 266:155785. [PMID: 39708520 DOI: 10.1016/j.prp.2024.155785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 12/08/2024] [Accepted: 12/18/2024] [Indexed: 12/23/2024]
Abstract
Several molecular strategies based on targeted gene delivery systems have been developed in recent years; however, the CRISPR-Cas9 technology introduced a new era of targeted gene editing, precisely modifying oncogenes, tumor suppressor genes, and other regulatory genes involved in carcinogenesis. However, efficiently and safely delivering CRISPR-Cas9 to cancer cells across the cell membrane and the nucleus is still challenging. Using viral vectors and nanoparticles presents issues of immunogenicity, off-target effects, and low targeting affinity. Naturally, extracellular vesicles called exosomes have garnered the most attention as delivery vehicles in oncology-related CRISPR-Cas9 calls due to their biocompatibility, loading capacity, and inherent targeting features. The following review discusses the current progress in using exosomes to deliver CRISPR-Cas9 components, the approaches to load the CRISPR components into exosomes, and the modification of exosomes to increase stability and tumor-targeted delivery. We discuss the latest strategies in targeting recently accomplished in the exosome field, including modifying the surface of exosomes to enhance their internalization by cancer cells, as well as the measures taken to overcome the impacts of TME on delivery efficiency. Focusing on in vitro and in vivo experimentation, this review shows that exosome-mediated CRISPR-Cas9 can potentially treat cancer types, including pancreatic, lymphoma, and leukemia, for given gene targets. This paper compares exosome-mediated delivery and conventional vectors regarding safety, immune response, and targeting ability. Last but not least, we present the major drawbacks and potential development of the seemingly promising field of exosome engineering in gene editing, with references to CRISPR technologies and applications that may help make the target exosomes therapeutic in oncology.
Collapse
Affiliation(s)
- Ashok Kumar Balaraman
- Research and Enterprise, University of Cyberjaya, Persiaran Bestari, Cyber 11, Cyberjaya, Selangor 63000, Malaysia
| | - M Arockia Babu
- Institute of Pharmaceutical Research, GLA UNIVERSITY, Mathura, UP 281406, India
| | - Ehssan Moglad
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al Kharj 11942, Saudi Arabia
| | - Viralkumar Mandaliya
- Marwadi University Research Center, Department of Microbiology, Faculty of Science, Marwadi University, Rajkot, Gujarat 360003, India
| | - M M Rekha
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Sofia Gupta
- Department of Chemistry, Chandigarh Engineering College, Chandigarh Group of Colleges-Jhanjeri, Mohali, Punjab 140307, India
| | - G V Siva Prasad
- Department of Chemistry, Raghu Engineering College, Visakhapatnam, Andhra Pradesh 531162, India
| | - Mukesh Kumari
- Department of Applied Sciences-Chemistry, NIMS Institute of Engineering & Technology, NIMS University Rajasthan, Jaipur, India
| | - Ashish Singh Chauhan
- Uttaranchal Institute of Pharmaceutical Sciences, Division of research and innovation, Uttaranchal University, Dehradun, Uttarakhand, India
| | - Haider Ali
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, India
| | - Kavita Goyal
- Department of Biotechnology, Graphic Era (Deemed to be University), Clement Town, Dehradun 248002, India.
| |
Collapse
|
46
|
Pavlov RV, Akimov SA, Dashinimaev EB, Bashkirov PV. Boosting Lipofection Efficiency Through Enhanced Membrane Fusion Mechanisms. Int J Mol Sci 2024; 25:13540. [PMID: 39769303 PMCID: PMC11677079 DOI: 10.3390/ijms252413540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 12/11/2024] [Accepted: 12/16/2024] [Indexed: 01/11/2025] Open
Abstract
Gene transfection is a fundamental technique in the fields of biological research and therapeutic innovation. Due to their biocompatibility and membrane-mimetic properties, lipid vectors serve as essential tools in transfection. The successful delivery of genetic material into the cytoplasm is contingent upon the fusion of the vector and cellular membranes, which enables hydrophilic polynucleic acids to traverse the hydrophobic barriers of two intervening membranes. This review examines the critical role of membrane fusion in lipofection efficiency, with a particular focus on the molecular mechanisms that govern lipoplex-membrane interactions. This analysis will examine the key challenges inherent to the fusion process, from achieving initial membrane proximity to facilitating final content release through membrane remodeling. In contrast to viral vectors, which utilize specialized fusion proteins, lipid vectors necessitate a strategic formulation and environmental optimization to enhance their fusogenicity. This review discusses recent advances in vector design and fusion-promoting strategies, emphasizing their potential to improve gene delivery yield. It highlights the importance of understanding lipoplex-membrane fusion mechanisms for developing next-generation delivery systems and emphasizes the need for continued fundamental research to advance lipid-mediated transfection technology.
Collapse
Affiliation(s)
- Rais V. Pavlov
- Research Institute for Systems Biology and Medicine, 18 Nauchniy Proezd, Moscow 117246, Russia
| | - Sergey A. Akimov
- Frumkin Institute of Physical Chemistry and Electrochemistry, Russian Academy of Sciences, 31/4 Leninskiy Prospekt, Moscow 119071, Russia;
| | - Erdem B. Dashinimaev
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Moscow 117997, Russia;
| | - Pavel V. Bashkirov
- Research Institute for Systems Biology and Medicine, 18 Nauchniy Proezd, Moscow 117246, Russia
| |
Collapse
|
47
|
Sun Y, He X, Han J, Yin W, Wang H, Li J, Liu W, Kuai X, Lv J, Ji J. Activated hepatic stellate cell-derived small extracellular vesicles facilitate M2 macrophage polarization and hepatoma progression via miR-27a-3p. Front Immunol 2024; 15:1489679. [PMID: 39742261 PMCID: PMC11685157 DOI: 10.3389/fimmu.2024.1489679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Accepted: 12/03/2024] [Indexed: 01/03/2025] Open
Abstract
The progression of hepatoma is heavily influenced by the microenvironment. Tumor-associated macrophages (TAMs) are considered to play a critical role in the tumor microenvironment (TME) and increase the aggressiveness of hepatoma. The activation of hepatic stellate cells (HSCs) is involved in hepatoma progression, and accumulating evidence demonstrates a change in microRNA (miRNA) expression during HSC activation. Therefore, the potential roles of HSCs-related miRNAs in macrophage differentiation and hepatoma progression deserve to be explored. The present study aimed to investigate the effects of miRNAs carried by small extracellular vesicles (sEVs) released by activated HSCs on hepatoma progression. The results indicated that miR-27a-3p was significantly upregulated in cells and corresponding sEVs during the activation of primary rat HSCs and human HSC line-LX2 cells. Furthermore, miR-27a-3p contributed to the proliferation and migration of hepatoma cells and promoted M2 polarization of macrophage. HSC-sEVs overexpressing miR-27a-3p can directly facilitate tumor progression and modulate macrophage polarization, indirectly contributing to hepatoma progression. Finally, Sprouty2 (SPRY2) was verified to be the target gene of miR-27a-3p. In conclusion, activated HSC-derived sEVs with high levels of miR-27a-3p might induce M2 macrophage polarization and promote hepatoma progression, providing new insights into the mechanism of hepatoma progression.
Collapse
Affiliation(s)
- Yufeng Sun
- Department of Pathology, Medical School of Nantong University, Nantong, China
- Key Laboratory of Microenvironment and Translational Cancer Research, Nantong, China
| | - Xiaoqian He
- Department of Pathology, Medical School of Nantong University, Nantong, China
- Key Laboratory of Microenvironment and Translational Cancer Research, Nantong, China
| | - Jiayi Han
- Department of Pathology, Medical School of Nantong University, Nantong, China
- Key Laboratory of Microenvironment and Translational Cancer Research, Nantong, China
| | - Wenxuan Yin
- Department of Pathology, Medical School of Nantong University, Nantong, China
| | - Haichen Wang
- Department of Pathology, Medical School of Nantong University, Nantong, China
| | - Jing Li
- Department of Pathology, Medical School of Nantong University, Nantong, China
| | - Weiqi Liu
- Department of Pathology, Medical School of Nantong University, Nantong, China
| | - Xingwang Kuai
- Department of Pathology, Medical School of Nantong University, Nantong, China
- Key Laboratory of Microenvironment and Translational Cancer Research, Nantong, China
| | - Jiaying Lv
- Department of Pathology, Medical School of Nantong University, Nantong, China
- Key Laboratory of Microenvironment and Translational Cancer Research, Nantong, China
| | - Juling Ji
- Department of Pathology, Medical School of Nantong University, Nantong, China
- Key Laboratory of Microenvironment and Translational Cancer Research, Nantong, China
| |
Collapse
|
48
|
Oliveira FD, Cavaco M, Figueira TN, Napoleão P, Valle J, Neves V, Andreu D, Castanho MA. vCPP2319 interacts with metastatic breast cancer extracellular vesicles (EVs) and transposes a human blood-brain barrier model. Heliyon 2024; 10:e40907. [PMID: 39717586 PMCID: PMC11664409 DOI: 10.1016/j.heliyon.2024.e40907] [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: 04/06/2024] [Revised: 12/02/2024] [Accepted: 12/03/2024] [Indexed: 12/25/2024] Open
Abstract
Brain metastases (BM) are frequently found in cancer patients and, though their precise incidence is difficult to estimate, there is evidence for a correlation between BM and specific primary cancers, such as lung, breast, and skin (melanoma). Among all these, breast cancer is the most frequently diagnosed among women and, in this case, BM cause a critical reduction of the overall survival (OS), especially in triple negative breast cancer (TNBC) patients. The main challenge of BM treatment is the impermeable nature of the blood-brain barrier (BBB), which shields the central nervous systems (CNS) from chemotherapeutic drugs. Extracellular vesicles (EVs) have been proposed as ideal natural drug carriers and these may exhibit some advantages over synthetic nanoparticles (NPs). In this work, we isolate breast cancer-derived EVs and study their ability to carry vCPP2319, a peptide with dual cell-penetration and anticancer activities. The selective cytotoxicity of anticancer peptide-loaded EVs towards breast cancer cells and their ability to translocate an in vitro BBB model are also addressed. Overall, it was possible to conclude that vCPP2319 naturally interacts with breast cancer-derived EVs, being retained at the surface of these vesicles. Moreover, the results revealed a cytotoxic activity for peptide-loaded EVs similar to that obtained with the peptide alone and the ability of peptide-loaded EVs to translocate an in vitro BBB model, which contrasts with the results obtained with the peptide alone. In conclusion, this work supports the use of EVs in the development of biological drug-delivery systems (DDS) capable of translocating the BBB.
Collapse
Affiliation(s)
- Filipa D. Oliveira
- Gulbenkian Institute for Molecular Medicine, Faculdade de Medicina da Universidade de Lisboa, Av. Prof. Egas Moniz, Lisbon, 1649-028, Portugal
| | - Marco Cavaco
- Gulbenkian Institute for Molecular Medicine, Faculdade de Medicina da Universidade de Lisboa, Av. Prof. Egas Moniz, Lisbon, 1649-028, Portugal
| | - Tiago N. Figueira
- Gulbenkian Institute for Molecular Medicine, Faculdade de Medicina da Universidade de Lisboa, Av. Prof. Egas Moniz, Lisbon, 1649-028, Portugal
| | - Patrícia Napoleão
- Gulbenkian Institute for Molecular Medicine, Faculdade de Medicina da Universidade de Lisboa, Av. Prof. Egas Moniz, Lisbon, 1649-028, Portugal
| | - Javier Valle
- Department of Medicine and Life Sciences, Pompeu Fabra University, Barcelona Biomedical Research Park, 08003, Barcelona, Spain
| | - Vera Neves
- Gulbenkian Institute for Molecular Medicine, Faculdade de Medicina da Universidade de Lisboa, Av. Prof. Egas Moniz, Lisbon, 1649-028, Portugal
| | - David Andreu
- Department of Medicine and Life Sciences, Pompeu Fabra University, Barcelona Biomedical Research Park, 08003, Barcelona, Spain
| | - Miguel A.R.B. Castanho
- Gulbenkian Institute for Molecular Medicine, Faculdade de Medicina da Universidade de Lisboa, Av. Prof. Egas Moniz, Lisbon, 1649-028, Portugal
| |
Collapse
|
49
|
Chen J, Jiang Y, Hou M, Liu C, Liu E, Zong Y, Wang X, Meng Z, Gu M, Su Y, Wang H, Fu J. Nuclear translocation of plasma membrane protein ADCY7 potentiates T cell-mediated antitumour immunity in HCC. Gut 2024; 74:128-140. [PMID: 39349007 DOI: 10.1136/gutjnl-2024-332902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 08/31/2024] [Indexed: 10/02/2024]
Abstract
BACKGROUND The potency of T cell-mediated responses is a determinant of immunotherapy effectiveness in treating malignancies; however, the clinical efficacy of T-cell therapies has been limited in hepatocellular carcinoma (HCC) owing to the extensive immunosuppressive microenvironment. OBJECTIVE Here, we aimed to investigate the key genes contributing to immune escape in HCC and raise a new therapeutic strategy for remoulding the HCC microenvironment. DESIGN The genome-wide in vivo clustered regularly interspaced short palindromic repeats (CRISPR) screen library was conducted to identify the key genes associated with immune tolerance. Single-cell RNA-seq (scRNA-seq), flow cytometry, HCC mouse models, chromatin immunoprecipitation and coimmunoprecipitation were used to explore the function and mechanism of adenylate cyclase 7 (ADCY7) in HCC immune surveillance. RESULTS Here, a genome-wide in vivo CRISPR screen identified a novel immune modulator-ADCY7. The transmembrane protein ADCY7 undergoes subcellular translocation via caveolae-mediated endocytosis and then translocates to the nucleus with the help of leucine-rich repeat-containing protein 59 (LRRC59) and karyopherin subunit beta 1 (KPNB1). In the nucleus, it functions as a transcription cofactor of CCAAT/enhancer binding protein alpha (CEBPA) to induce CCL5 transcription, thereby increasing CD8+ T cell infiltration to restrain HCC progression. Furthermore, ADCY7 can be secreted as exosomes and enter neighbouring tumour cells to promote CCL5 induction. Exosomes with high ADCY7 levels promote intratumoural infiltration of CD8+ T cells and suppress HCC tumour growth. CONCLUSION We delineate the unconventional function and subcellular location of ADCY7, highlighting its pivotal role in T cell-mediated immunity in HCC and its potential as a promising treatment target.
Collapse
Affiliation(s)
- Jianan Chen
- International Cooperation Laboratory on Signal Transduction, National Center for Liver Cancer, Ministry of Education Key Laboratory on Signaling Regulation and Targeting Therapy of Liver Cancer, Shanghai Key Laboratory of Hepato-biliary Tumor Biology, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, China
| | - Youhai Jiang
- Cancer Research Center, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology, Anhui, China
| | - Minghui Hou
- Research Center for Organoids, Department of Translational Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Chunliang Liu
- International Cooperation Laboratory on Signal Transduction, National Center for Liver Cancer, Ministry of Education Key Laboratory on Signaling Regulation and Targeting Therapy of Liver Cancer, Shanghai Key Laboratory of Hepato-biliary Tumor Biology, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, China
| | - Erdong Liu
- Institute of Metabolism & Integrative Biology, Fudan University, Shanghai, China
| | - Yali Zong
- Institute of Metabolism & Integrative Biology, Fudan University, Shanghai, China
| | - Xiang Wang
- Second Department of Biliary Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, China
| | - Zhengyuan Meng
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mingye Gu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yu Su
- International Cooperation Laboratory on Signal Transduction, National Center for Liver Cancer, Ministry of Education Key Laboratory on Signaling Regulation and Targeting Therapy of Liver Cancer, Shanghai Key Laboratory of Hepato-biliary Tumor Biology, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, China
| | - Hongyang Wang
- International Cooperation Laboratory on Signal Transduction, National Center for Liver Cancer, Ministry of Education Key Laboratory on Signaling Regulation and Targeting Therapy of Liver Cancer, Shanghai Key Laboratory of Hepato-biliary Tumor Biology, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, China
| | - Jing Fu
- International Cooperation Laboratory on Signal Transduction, National Center for Liver Cancer, Ministry of Education Key Laboratory on Signaling Regulation and Targeting Therapy of Liver Cancer, Shanghai Key Laboratory of Hepato-biliary Tumor Biology, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, China
| |
Collapse
|
50
|
Chen Z, Li X, Liu Q, Li W, Wang T, Ding D. Recent advances in AIE-based platforms for cancer immunotherapy. J Control Release 2024; 376:1-19. [PMID: 39343142 DOI: 10.1016/j.jconrel.2024.09.041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Revised: 09/11/2024] [Accepted: 09/24/2024] [Indexed: 10/01/2024]
Abstract
Aggregation-induced emission luminogens (AIEgens) possess the unique property of enhanced fluorescence and photostability in aggregated states, making them exceptional materials for the convergence of imaging and phototherapy. With their inherent advantages, AIEgens are propelling the field of nanomedicine into a vibrant frontier in the phototheranostics of a spectrum of diseases, particularly in the realm of cancer immunotherapy. AIEgens-based therapeutics enhance the cancer immune response through a variety of approaches, including real-time image-guided precise therapy, induction of programmed cell death, metabolic reprogramming, and modulation of the tumor microenvironment. Additionally, they contribute to the synergistic effect of immune checkpoint inhibition, a pivotal aspect of modern cancer immunotherapy strategies. This review offers a comprehensive overview of the integration of AIEgens in nanomedicine and their role in immune adaptation, highlighting the advantages, basic action mechanisms, and recent advancement of AIEgens as promising therapeutic platform for cancer immunotherapy.
Collapse
Affiliation(s)
- Ziyi Chen
- Frontiers Science Center for New Organic Matter, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, and College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Xueping Li
- Tianjin Key Laboratory of Biomedical Materials and Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China
| | - Qian Liu
- Department of Urology, Tianjin First Central Hospital, Tianjin 300192, China
| | - Wen Li
- Tianjin Key Laboratory of Biomedical Materials and Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China.
| | - Tianjiao Wang
- Tianjin Key Laboratory of Biomedical Materials and Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China.
| | - Dan Ding
- Frontiers Science Center for New Organic Matter, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, and College of Life Sciences, Nankai University, Tianjin 300071, China; Nankai International Advanced Research Institute, Futian District, Shenzhen 518045, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou 221002, Jiangsu, China.
| |
Collapse
|