1
|
Daghrery A, Araújo IJDS, Marques JF, Alipour M, Ünsal RBK, Chathoth BM, Sivaramakrishnan G, Delgadillo-Barrera S, Chaurasia A. Role of exosomes in dental and craniofacial regeneration - A review. Tissue Cell 2025; 93:102684. [PMID: 39740273 DOI: 10.1016/j.tice.2024.102684] [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: 08/16/2024] [Revised: 12/05/2024] [Accepted: 12/11/2024] [Indexed: 01/02/2025]
Abstract
BACKGROUND The treatment of congenital deformities, traumatic injuries, infectious diseases, and tumors in the craniomaxillofacial (CMF) region is complex due to the intricate nature of the tissues involved. Conventional treatments such as bone grafts and cell transplantation face limitations, including the need for multiple surgeries, complications, and safety concerns. OBJECTIVE This paper aims to provide a comprehensive analysis of the role of exosomes (EXOs) in CMF and dental tissue regeneration and to explore their potential applications in regenerative dental medicine. METHODS An extensive review of advancements in tissue engineering, materials sciences, and nanotechnology was conducted to evaluate the development of delivery systems for EXOs-based therapies. The analysis included how EXOs, as nanovesicles released by cells, can be modified to target specific cells or loaded with functional molecules for drug or gene delivery. RESULTS EXOs have emerged as a promising alternative to cell transplant therapy, offering a safer method for cell communication and epigenetic control. EXOs transport important proteins and genetic materials, facilitating intercellular communication and delivering therapeutics effectively. The potential of EXOs in personalized medicine, particularly in diagnosing, customizing treatment, and predicting patient responses, is highlighted. CONCLUSION EXO-mediated therapy holds significant potential for advancing tissue regeneration, offering targeted, personalized treatment options with reduced side effects. However, challenges in purification, production, and standardized protocols need to be addressed before its clinical application can be fully realized.
Collapse
Affiliation(s)
- Arwa Daghrery
- Department of Restorative Dental Sciences, School of Dentistry, Jazan University, Jazan, Kingdom of Saudi Arabia.
| | | | - Joana Faria Marques
- Faculdade de Medicina Dentária, Universidade de Lisboa, Cidade Universitária, Lisboa 1600-277, Portugal.
| | - Mahdieh Alipour
- Dental and Periodontal Research Center, Faculty of Dentistry, Tabriz University of Medical Sciences, Iran; Departments of Oral and Craniofacial Sciences, School of Dental Medicine, University of Pittsburgh, USA.
| | - Revan Birke Koca Ünsal
- Department of Periodontology, University of Kyrenia, Faculty of Dentistry, Kyrenia, Cyprus.
| | | | | | - Sara Delgadillo-Barrera
- Grupo de Investigacion Básica y Aplicada en Odontología - IBAPO, Facultad de Odontologia, Universidad Nacional de Colombia, Bogotá, Colombia.
| | - Akhilanand Chaurasia
- Department of Oral Medicine and Radiology, Faculty of Dental Sciences. King George's Medical University, Lucknow, India.
| |
Collapse
|
2
|
Wu Q, Chen S, Xie X, Yan H, Feng X, Su G, Zhang Z. Glioblastoma- derived exosomes (GBM-Exo) regulate microglial M2 polarization via the RAC1/AKT/NRF2 pathway. J Neurooncol 2025; 172:447-460. [PMID: 40019712 DOI: 10.1007/s11060-024-04934-6] [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: 11/21/2024] [Accepted: 12/27/2024] [Indexed: 03/01/2025]
Abstract
PURPOSE The impact of exosome-mediated communication between glioblastoma and microglia on the formation of an immunosuppressive microenvironment remains to be explored. Tumor-associated macrophages are more likely to adopt an M2-like phenotype within the immunosuppressive environment. Here, we investigate the molecular mechanisms by which glioblastoma-derived exosomes promote microglial M2 polarization through RAC1. METHODS The expression of RAC1 in GBM was collected from public databases. A C57BL/6 mouse glioma xenograft model was established using intracranial stereotactic injection. RAC1 expression was validated by qRT-PCR, Western blotting, and immunohistochemistry. Glioblastoma-derived exosomes were isolated by ultracentrifugation and characterized by Nanoparticle Tracking Analysis (NTA), transmission electron microscopy, and Western blotting for exosome markers, with the content of RAC1 being profiled. RAC1 and AKT inhibitors were used to co-treat microglia with exosomes. Microglial polarization under different treatment conditions was assessed by Western blotting and immunofluorescence. RESULT Our study reveals that RAC1 is aberrantly expressed in glioblastoma and is associated with macrophage immune infiltration. GBM-derived exosomes, carrying RAC1, promote the M2 polarization of microglia. In microglia treated with GBM-derived exosomes, inhibition of RAC1 activity suppressed AKT phosphorylation and NRF2 nuclear translocation, while reducing the expression of M2 phenotype markers. Notably, following AKT inhibition, the exosome-induced NRF2 nuclear translocation was also significantly suppressed, highlighting the critical role of RAC1-mediated AKT activation in NRF2 translocation and microglial M2 polarization. CONCLUSION Our study demonstrates that RAC1-carrying GBM-exosomes promote M2 polarization of microglia, a process mediated through the RAC1/AKT/NRF2 pathway.
Collapse
Affiliation(s)
- Qionghui Wu
- Lanzhou University Second Hospital, Lanzhou University, Lanzhou, Gansu Province, 730000, China
| | - Shanlin Chen
- Department of Neurosurgery, Liangzhou Hospital of Wuwei, Wuwei, Gansu Province, 733000, China
| | - Xiaodong Xie
- Institute of Genetics, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu Province, 730000, China
| | - Hong Yan
- Lanzhou University Second Hospital, Lanzhou University, Lanzhou, Gansu Province, 730000, China
| | - Xinli Feng
- Lanzhou University Second Hospital, Lanzhou University, Lanzhou, Gansu Province, 730000, China
| | - Gang Su
- Institute of Genetics, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu Province, 730000, China.
| | - Zhenchang Zhang
- Lanzhou University Second Hospital, Lanzhou University, Lanzhou, Gansu Province, 730000, China.
| |
Collapse
|
3
|
Mukerjee N, Bhattacharya A, Maitra S, Kaur M, Ganesan S, Mishra S, Ashraf A, Rizwan M, Kesari KK, Tabish TA, Thorat ND. Exosome isolation and characterization for advanced diagnostic and therapeutic applications. Mater Today Bio 2025; 31:101613. [PMID: 40161926 PMCID: PMC11950786 DOI: 10.1016/j.mtbio.2025.101613] [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: 09/22/2024] [Revised: 02/01/2025] [Accepted: 02/24/2025] [Indexed: 04/02/2025] Open
Abstract
Advancements in exosome isolation technologies are pivotal for transforming personalized medicine and enhancing clinical diagnostics. Exosomes, small extracellular vesicles with diameters ranging between 30 and 150 nm, are secreted into bodily fluids by a variety of cells and play essential roles in intercellular communication. These vesicles facilitate the transfer of nucleic acids, lipids, and proteins, affecting a wide range of biological and pathological processes. Given their importance in disease diagnostics, therapy, and as biomarkers, there has been a surge in developing methods to isolate them from fluids such as urine, saliva, blood, and cerebrospinal fluid. While traditional isolation techniques like ultracentrifugation and polymer-based precipitation have been foundational, recent technological advances have introduced more precise methods like microfluidics and immunoaffinity capture. These newer methods enable high-throughput and specific exosome isolation by targeting surface markers, thus enhancing purity. However, challenges such as balancing purity with yield and the lack of standardized protocols across different laboratories persist, impacting the consistency of findings. By integrating advanced isolation techniques and discussing their implications in diagnostics and therapy, this review aims to catalyze further research and adoption of exosome-based technologies in medicine, marking a significant stride towards tailored healthcare solutions.
Collapse
Affiliation(s)
- Nobendu Mukerjee
- Centre for Infectious Diseases & Microbiology, School of Public Health Sciences and Technology, Malla Reddy Vishwavidyapeeth, Hyderabad 500 055, Telangana, India
| | - Arghya Bhattacharya
- Department of Pharmacology, Bengal School of Technology, West Bengal, Kolkata, 712102, India
| | - Swastika Maitra
- Center for Global Health Research, Saveetha Medical College and Hospital, Saveetha Institute of Medical and Technical Sciences, Chennai, India
| | - Mandeep Kaur
- Department of Sciences, Vivekananda Global University, Jaipur, Rajasthan, 303012, India
| | - Subbulakshmi Ganesan
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Shivang Mishra
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, India
| | - Ayash Ashraf
- Chandigarh Pharmacy College, Chandigarh Group of College, Jhanjeri, Mohali, 140307, Punjab, India
| | - Muhammad Rizwan
- Department of Biomedical Engineering, Department of Ophthalmology, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | | | - Tanveer A. Tabish
- Radcliffe Department of Medicine, University of Oxford, OX3 7BN, United Kingdom
| | - Nanasaheb D. Thorat
- Department of Physics and Bernal Institute, University of Limerick, Castletroy, Limerick V94T9PX, Ireland
- Limerick Digital Cancer Research Centre (LDCRC) University of Limerick, Castletroy, Limerick, V94T9PX, Ireland
| |
Collapse
|
4
|
Saadh MJ, Hussain QM, Alazzawi TS, Fahdil AA, Athab ZH, Yarmukhamedov B, Al-Nuaimi AMA, Alsaikhan F, Farhood B. MicroRNA as Key Players in Hepatocellular Carcinoma: Insights into Their Role in Metastasis. Biochem Genet 2025; 63:1014-1062. [PMID: 39103713 DOI: 10.1007/s10528-024-10897-0] [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: 06/13/2024] [Accepted: 07/29/2024] [Indexed: 08/07/2024]
Abstract
Liver cancer or hepatocellular carcinoma (HCC) remains the most common cancer in global epidemiology. Both the frequency and fatality of this malignancy have shown an upward trend over recent decades. Liver cancer is a significant concern due to its propensity for both intrahepatic and extrahepatic metastasis. Liver cancer metastasis is a multifaceted process characterized by cell detachment from the bulk tumor, modulation of cellular motility and invasiveness, enhanced proliferation, avoidance of the immune system, and spread either via lymphatic or blood vessels. MicroRNAs (miRNAs) are small non-coding ribonucleic acids (RNAs) playing a crucial function in the intricate mechanisms of tumor metastasis. A number of miRNAs can either increase or reduce metastasis via several mechanisms, such as control of motility, proliferation, attack by the immune system, cancer stem cell properties, altering the microenvironment, and the epithelial-mesenchymal transition (EMT). Besides, two other types of non-coding RNAs, such as long non-coding RNAs (lncRNAs) and circular RNAs (circRNAs) can competitively bind to endogenous miRNAs. This competition results in the impaired ability of the miRNAs to inhibit the expression of the specific messenger RNAs (mRNAs) that are targeted. Increasing evidence has shown that the regulatory axis comprising circRNA/lncRNA-miRNA-mRNA is correlated with the regulation of HCC metastasis. This review seeks to present a thorough summary of recent research on miRNAs in HCC, and their roles in the cellular processes of EMT, invasion and migration, as well as the metastasis of malignant cells. Finally, we discuss the function of the lncRNA/circRNA-miRNA-mRNA network as a crucial modulator of carcinogenesis and the regulation of signaling pathways or genes that are relevant to the metastasis of HCC. These findings have the potential to offer valuable insight into the discovery of novel therapeutic approaches for management of liver cancer metastasis.
Collapse
Affiliation(s)
- Mohamed J Saadh
- Faculty of Pharmacy, Middle East University, Amman, 11831, Jordan
| | | | - Tuqa S Alazzawi
- College of Dentist, National University of Science and Technology, Nasiriyah, Dhi Qar, Iraq
| | - Ali A Fahdil
- Medical Technical College, Al-Farahidi University, Baghdad, Iraq
| | - Zainab H Athab
- Department of Pharmacy, Al-Zahrawi University College, Karbala, Iraq
| | - Bekhzod Yarmukhamedov
- Department of Public Health and Healthcare management, Samarkand State Medical University, 18 Amir Temur Street, Samarkand, Uzbekistan
| | | | - Fahad Alsaikhan
- College of Pharmacy, Prince Sattam Bin Abdulaziz University, Alkharj, Saudi Arabia.
- School of Pharmacy, Ibn Sina National College for Medical Studies, Jeddah, Saudi Arabia.
| | - Bagher Farhood
- Department of Medical Physics and Radiology, Faculty of Paramedical Sciences, Kashan University of Medical Sciences, Kashan, Iran.
| |
Collapse
|
5
|
Feng G, Lan X, Qin S, Shi Y, Zhao Q, Li Q, Zhong L. Advances in Research on Exosomal miRNAs in Central Nervous System Diseases. ASN Neuro 2025; 17:2465546. [PMID: 40165664 DOI: 10.1080/17590914.2025.2465546] [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: 10/29/2024] [Revised: 01/03/2025] [Accepted: 02/03/2025] [Indexed: 04/02/2025] Open
Abstract
Neurological diseases present a wide range of conditions, intricate diagnosis and treatment processes, and complex prognostic considerations. Therefore, research focusing on the diagnosis and treatment of these diseases is crucial. Exosomal miRNAs are small RNA molecules enclosed in membrane vesicles, released by cells and known to play roles in the development of various neurological disorders. They also serve as specific biomarkers for these conditions. Drawing on extensive research on exosomal miRNAs in diseases like stroke, Alzheimer's, epilepsy, Parkinson's, and neuroregeneration, this paper provides a comprehensive review of the relationship between exosomal miRNAs and neurological diseases. We strive to offer current and detailed theoretical understandings to help with the diagnosis and treatment of these disorders.
Collapse
Affiliation(s)
- Guangli Feng
- The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Xiaoqian Lan
- The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Shiyi Qin
- The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Yuting Shi
- Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Qinxi Zhao
- The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Qing Li
- Yunnan Key Laboratory of Stem Cell and Regenerative Medicine, Kunming Medical University, Kunming, Yunnan, China
| | - Lianmei Zhong
- Xuanwu Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
6
|
Ge W, Mu Z, Yang S, Zeng Y, Deng Y, Lin Y, Xie P, Li G. Biosensor-based methods for exosome detection with applications to disease diagnosis. Biosens Bioelectron 2025; 279:117362. [PMID: 40157151 DOI: 10.1016/j.bios.2025.117362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 02/09/2025] [Accepted: 03/09/2025] [Indexed: 04/01/2025]
Abstract
Exosomes are nanoscale extracellular vesicles (EVs) secreted by most eukaryotic cells and can be found in nearly all human body fluids. Increasing evidence has revealed their pivotal roles in intercellular communication, and their active participation in myriad physiological and pathological activities. Exosomes' functions rely on their contents that are closely correlated with the biological characteristics of parental cells, which may provide a rich resource of molecular information for accurate and detailed diagnosis of a diverse array of diseases, such as differential diagnosis of Alzheimer's disease, early detection and subtyping of various tumors. As a category of sensitive detection devices, biosensors can fully reveal the molecular information and convert them into actionable clinical information. In this review, recent advances in biosensor-based methods for the detection of exosomes are summarized. We have described the fabrication of various biosensors based on the analysis of exosomal proteins, RNAs or glycans for accurate diagnosis, with respect to their elaborate recognition designs, signal amplification strategies, sensing properties, as well as their application potential. The challenges along with corresponding technologies in the future development and clinical translation of these biosensors are also discussed.
Collapse
Affiliation(s)
- Weikang Ge
- State Key Laboratory of Analytical Chemistry for Life Science, School of Life Sciences, Nanjing University, Nanjing 210023, People's Republic of China
| | - Zheying Mu
- State Key Laboratory of Analytical Chemistry for Life Science, School of Life Sciences, Nanjing University, Nanjing 210023, People's Republic of China
| | - Shiao Yang
- State Key Laboratory of Analytical Chemistry for Life Science, School of Life Sciences, Nanjing University, Nanjing 210023, People's Republic of China
| | - Yujing Zeng
- Center for Molecular Recognition and Biosensing, School of Life Sciences, Shanghai University, Shanghai 200444, People's Republic of China
| | - Ying Deng
- State Key Laboratory of Analytical Chemistry for Life Science, School of Life Sciences, Nanjing University, Nanjing 210023, People's Republic of China
| | - Yifan Lin
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, People's Republic of China
| | - Ping Xie
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, People's Republic of China.
| | - Genxi Li
- State Key Laboratory of Analytical Chemistry for Life Science, School of Life Sciences, Nanjing University, Nanjing 210023, People's Republic of China; Center for Molecular Recognition and Biosensing, School of Life Sciences, Shanghai University, Shanghai 200444, People's Republic of China.
| |
Collapse
|
7
|
Wu YY, Yu LSY, Zhou HY, Xue JC. Effect of HepG2-Derived Exosome with PDGF-D Knockdown on Transformation of Normal Fibroblasts into Tumor-Associated Fibroblasts in Liver Cancer. FRONT BIOSCI-LANDMRK 2025; 30:26045. [PMID: 40152369 DOI: 10.31083/fbl26045] [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: 08/07/2024] [Revised: 09/12/2024] [Accepted: 09/26/2024] [Indexed: 03/29/2025]
Abstract
BACKGROUND It is known that the transformation of liver cancer-mediated fibroblasts into cancer-related fibroblasts (CAFs) is beneficial to the development of liver cancer. However, the specific mechanism is still unclear. METHODS Human hepatocarcinoma (HepG2) cells were treated with short hairpin RNA (shRNA) of platelet-derived growth factor-D (shPDGF-D) vector, and the exosomes secreted by the cells were separated using ultracentrifugation and identified by using nanoparticle tracking analysis, transmission electron microscope, and western blot analysis. Exosomes were co-cultured with mouse primary fibroblasts, and then the activity, proliferation, cell cycle, migration, epithelial-mesenchymal transition- (EMT-) and CAF marker-related protein expression levels of fibroblasts were determined by cell counting kit-8 (CCK-8), immunofluorescence, flow cytometry, wound healing, real-time reverse transcription-PCR, and western blotting assays, respectively. Co-cultured fibroblasts were mixed with HepG2 cells and injected subcutaneously into mice to construct animal models. The size and weight of xenograft tumor and the expression of epithelial-mesenchymal transition- (EMT-), angiogenesis- and CAFs marker-related proteins were detected. RESULTS The exosomes inhibited the proliferation, migration, EMT, and induced cell cycle arrest, as well as decreased the expression of α-SMA, FAP, MMP-9, and VEGF in fibroblasts. In vivo, sh-PDGF-D inhibited tumor growth, reduced the expressions of CD31, vimentin, α-SMA, FAP, MMP9, and VEGF, and promoted the expression of E-cadherin. CONCLUSIONS Exosomes derived from HepG2 cells transfected with shPDGF-D prevent normal fibroblasts from transforming into CAFs, thus inhibiting angiogenesis and EMT of liver cancer.
Collapse
Affiliation(s)
- Yan-Yan Wu
- Department of Pharmacy, Tongde Hospital of Zhejiang Province, 310012 Hangzhou, Zhejiang, China
| | - Liu-Shen-Yan Yu
- Department of Pharmacy, Tongde Hospital of Zhejiang Province, 310012 Hangzhou, Zhejiang, China
| | - Han-Yu Zhou
- Department of Pharmacy, Tongde Hospital of Zhejiang Province, 310012 Hangzhou, Zhejiang, China
| | - Jun-Chao Xue
- Department of Pharmacy, Tongde Hospital of Zhejiang Province, 310012 Hangzhou, Zhejiang, China
| |
Collapse
|
8
|
Azar BKY, Vakhshiteh F. The Pre-metastatic Niche: How Cancer Stem Cell-Derived Exosomal MicroRNA Fit into the Puzzle. Stem Cell Rev Rep 2025:10.1007/s12015-025-10866-z. [PMID: 40095238 DOI: 10.1007/s12015-025-10866-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/12/2025] [Indexed: 03/19/2025]
Abstract
Cancer metastasis is a complicated biological process that critically affects cancer progression, patient outcomes, and treatment plans. A significant step in metastasis is the formation of a pre-metastatic niche (PMN). A small subset of cells within tumors, known as cancer stem cells (CSCs), possess unique characteristics including, differentiation into different cell types within the tumor, self-renewal, and resistance to conventional therapies, that enable them to initiate tumors and drive metastasis. PMN plays an important role in preparing secondary organs for the arrival and proliferation of CSCs, thereby facilitating metastasis. CSC-derived exosomes are crucial components in the complex interplay between CSCs and the tumor microenvironment. These exosomes function as transporters of various substances that can promote cancer progression, metastasis, and modulation of pre-metastatic environments by delivering microRNA (miRNA, miR) cargo. This review aims to illustrate how exosomal miRNAs (exo-miRs) secreted by CSCs can predispose PMN and promote angiogenesis and metastasis.
Collapse
Affiliation(s)
- Behjat Kheiri Yeghaneh Azar
- Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Faezeh Vakhshiteh
- Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran.
| |
Collapse
|
9
|
Zhang X, Artz N, Steindler DA, Hingtgen S, Satterlee AB. Exosomes: Traversing the blood-brain barrier and their therapeutic potential in brain cancer. Biochim Biophys Acta Rev Cancer 2025; 1880:189300. [PMID: 40097050 DOI: 10.1016/j.bbcan.2025.189300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 03/07/2025] [Accepted: 03/09/2025] [Indexed: 03/19/2025]
Abstract
The blood-brain barrier (BBB) presents a major challenge for the effective delivery of therapeutic agents to the brain tumor cells from the peripheral blood circulation, making the treatment of central nervous system (CNS)-related cancers more difficult and resistant to both standard treatments and emerging therapies. Exosomes, which serve as messengers for intercellular communication throughout the body, can naturally or be modified to penetrate the BBB. Recently, exosomes have been increasingly explored as an invasive or non-invasive approach for delivering therapeutic agents to the CNS. With their low immunogenicity, ease of modification, excellent cargo protection, and inherent ability to cross the BBB, exosomes hold great promise for revolutionizing targeted therapy for CNS-related diseases, including brain cancer. In this review, we highlight recent discoveries and insights into the mechanisms exosomes use to penetrate the BBB, the methods they employ to payload diverse therapeutics, and their roles in transporting therapeutic compounds for brain cancer and other neurological disorders.
Collapse
Affiliation(s)
- Xiaopei Zhang
- Eshelman School of Pharmacy, Division of Pharmacoengineering and Molecular Pharmaceutics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| | - Nichole Artz
- Department of Pediatric Hematology/Oncology, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Dennis A Steindler
- Steindler Consulting, Boston, MA, USA; Eshelman Institute for Innovation, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| | - Shawn Hingtgen
- Eshelman School of Pharmacy, Division of Pharmacoengineering and Molecular Pharmaceutics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Andrew Benson Satterlee
- Eshelman School of Pharmacy, Division of Pharmacoengineering and Molecular Pharmaceutics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Eshelman Institute for Innovation, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
10
|
Geng JX, Lu YF, Zhou JN, Huang B, Qin Y. Exosome technology: A novel and effective drug delivery system in the field of cancer therapy. World J Gastrointest Oncol 2025; 17:101857. [PMID: 40092946 PMCID: PMC11866225 DOI: 10.4251/wjgo.v17.i3.101857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Revised: 11/23/2024] [Accepted: 12/20/2024] [Indexed: 02/14/2025] Open
Abstract
In this article, we revisit an article, which specifically focuses on the utilization of exosomes derived from human bone marrow mesenchymal stem cells (MSCs) for targeted delivery of gemcitabine in pancreatic cancer treatment. The experimental results demonstrated that the exosome-based drug delivery system derived from MSCs significantly augmented apoptosis in pancreatic cancer cells. The biocompatibility, targeting specificity, and low immunogenicity of exosomes render them as optimal carriers for drug delivery, enabling precise administration of therapeutics to diseased tissues while mitigating adverse effects, thereby achieving targeted treatment of cancer cells and significantly enhancing anti-tumor efficacy. However, the clinical application of exosome drug delivery platforms in oncology still presents challenges, necessitating further optimization to ensure their stability and efficacy. This study focuses on elucidating the advantages of exosomes as a drug delivery platform, exploring the utilization of MSC-derived exosomes in oncology therapy, and discussing their potential and future directions in cancer treatment.
Collapse
Affiliation(s)
- Jia-Xin Geng
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, Zhejiang Province, China
| | - Yao-Fan Lu
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, Zhejiang Province, China
| | - Jing-Nan Zhou
- Zhejiang Cancer Hospital, Hangzhou 310018, Zhejiang Province, China
| | - Biao Huang
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, Zhejiang Province, China
| | - Yuan Qin
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, Zhejiang Province, China
| |
Collapse
|
11
|
McAtee C, Patel M, Hoshino D, Sung BH, von Lersner A, Shi M, Hong NH, Young A, Krystofiak E, Zijlstra A, Weaver AM. Secreted exosomes induce filopodia formation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.07.20.604139. [PMID: 40161676 PMCID: PMC11952364 DOI: 10.1101/2024.07.20.604139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Filopodia are dynamic adhesive cytoskeletal structures that are critical for directional sensing, polarization, cell-cell adhesion, and migration of diverse cell types. Filopodia are also critical for neuronal synapse formation. While dynamic rearrangement of the actin cytoskeleton is known to be critical for filopodia biogenesis, little is known about the upstream extracellular signals. Here, we identify secreted exosomes as potent regulators of filopodia formation. Inhibition of exosome secretion inhibited the formation and stabilization of filopodia in both cancer cells and neurons and inhibited subsequent synapse formation by neurons. Rescue experiments with purified small and large extracellular vesicles (EVs) identified exosome-enriched small EVs (SEVs) as having potent filopodia-inducing activity. Proteomic analyses of cancer cell-derived SEVs identified the TGF-β family coreceptor endoglin as a key SEV-enriched cargo that regulates filopodia. Cancer cell endoglin levels also affected filopodia-dependent behaviors, including metastasis of cancer cells in chick embryos and 3D migration in collagen gels. As neurons do not express endoglin, we performed a second proteomics experiment to identify SEV cargoes regulated by endoglin that might promote filopodia in both cell types. We discovered a single SEV cargo that was altered in endoglin-KD cancer SEVs, the transmembrane protein Thrombospondin Type 1 Domain Containing 7A (THSD7A). We further found that both cancer cell and neuronal SEVs carry THSD7A and that add-back of purified THSD7A is sufficient to rescue filopodia defects of both endoglin-KD cancer cells and exosome-inhibited neurons. We also find that THSD7A induces filopodia formation through activation of the Rho GTPase, Cdc42. These findings suggest a new model for filopodia formation, triggered by exosomes carrying THSD7A.
Collapse
|
12
|
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 PMCID: PMC11940788 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
|
13
|
Wang D, Shen J, Wang Y, Cui H, Li Y, Zhou L, Li G, Wang Q, Feng X, Qin M, Dong B, Yang P, Li Y, Ma X, Ma J. Mechanisms of Ferroptosis in bone disease: A new target for osteoporosis treatment. Cell Signal 2025; 127:111598. [PMID: 39788305 DOI: 10.1016/j.cellsig.2025.111598] [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: 11/15/2024] [Revised: 12/23/2024] [Accepted: 01/06/2025] [Indexed: 01/12/2025]
Abstract
Osteoporosis (OP) is a common disease in the elderly, characterized by decreased bone strength, reduced bone density, and increased fracture risk. There are two clinical types of osteoporosis: primary osteoporosis and secondary osteoporosis. The most common form is postmenopausal osteoporosis, which is caused by decreased estrogen production after menopause. Secondary osteoporosis, on the other hand, occurs when certain medications, diabetes, or nutritional deficiencies lead to a decrease in bone density. Ferroptosis, a new iron-dependent programmed cell death process, is critical in regulating the development of osteoporosis, but the underlying molecular mechanisms are complex. In the pathologic process of osteoporosis, several studies have found that ferroptosis may occur in osteocytes, osteoblasts, and osteoclasts, cell types closely related to bone metabolism. The imbalance of iron homeostasis in osteoblasts and excessive iron accumulation can promote lipid peroxidation through the Fenton reaction, which induces ferroptosis in osteoblasts and affects their role in regulating bone metabolism. Ferroptosis in osteoblasts inhibits bone formation and reduces the amount of new bone production. Osteoclast-associated ferroptosis abnormalities, on the other hand, may alter the homeostasis of bone resorption. In this paper, we start from the molecular mechanism of ferroptosis, and introduce the ways in which ferroptosis affects the physiological and pathological processes of the body. After that, the effects of ferroptosis on osteoblasts and osteoclasts will be discussed separately to elucidate the molecular mechanism between ferroptosis and osteoporosis, which will provide a new breakthrough for the prevention and treatment of osteoporosis and a more effective and better idea for the treatment strategy of osteoporosis.
Collapse
Affiliation(s)
- Dong Wang
- College of Integrative Medicine of Tianjin University of traditional Chinese Medicine, Tianjin 301617,China; Graduate School of Tianjin University of traditional Chinese Medicine, Tianjin 301617, China
| | - Jiahui Shen
- College of Integrative Medicine of Tianjin University of traditional Chinese Medicine, Tianjin 301617,China; Graduate School of Tianjin University of traditional Chinese Medicine, Tianjin 301617, China
| | - Yan Wang
- Tianjin Hospital of Tianjin University (Tianjin Hospital), Tianjin 300211, China; Tianjin Orthopedic Institute, Tianjin 300050, China; Tianjin Key Laboratory of Orthopedic Biomechanics and Medical Engineering, Tianjin 300050, China
| | - Hongwei Cui
- Tianjin Hospital of Tianjin University (Tianjin Hospital), Tianjin 300211, China; Tianjin Orthopedic Institute, Tianjin 300050, China; Tianjin Key Laboratory of Orthopedic Biomechanics and Medical Engineering, Tianjin 300050, China
| | - Yanxin Li
- Tianjin Hospital of Tianjin University (Tianjin Hospital), Tianjin 300211, China; Tianjin Orthopedic Institute, Tianjin 300050, China; Tianjin Key Laboratory of Orthopedic Biomechanics and Medical Engineering, Tianjin 300050, China
| | - Liyun Zhou
- Tianjin Hospital of Tianjin University (Tianjin Hospital), Tianjin 300211, China; Tianjin Orthopedic Institute, Tianjin 300050, China; Tianjin Key Laboratory of Orthopedic Biomechanics and Medical Engineering, Tianjin 300050, China
| | - Guang Li
- Tianjin Hospital of Tianjin University (Tianjin Hospital), Tianjin 300211, China; Tianjin Orthopedic Institute, Tianjin 300050, China; Tianjin Key Laboratory of Orthopedic Biomechanics and Medical Engineering, Tianjin 300050, China
| | - Qiyu Wang
- College of Integrative Medicine of Tianjin University of traditional Chinese Medicine, Tianjin 301617,China; Graduate School of Tianjin University of traditional Chinese Medicine, Tianjin 301617, China
| | - Xiaotian Feng
- College of Integrative Medicine of Tianjin University of traditional Chinese Medicine, Tianjin 301617,China; Graduate School of Tianjin University of traditional Chinese Medicine, Tianjin 301617, China
| | - Mengran Qin
- Tianjin Hospital of Tianjin University (Tianjin Hospital), Tianjin 300211, China; Tianjin Orthopedic Institute, Tianjin 300050, China; Tianjin Key Laboratory of Orthopedic Biomechanics and Medical Engineering, Tianjin 300050, China
| | - Benchao Dong
- Tianjin Hospital of Tianjin University (Tianjin Hospital), Tianjin 300211, China; Tianjin Orthopedic Institute, Tianjin 300050, China; Tianjin Key Laboratory of Orthopedic Biomechanics and Medical Engineering, Tianjin 300050, China
| | - Peichuan Yang
- Tianjin Hospital of Tianjin University (Tianjin Hospital), Tianjin 300211, China; Tianjin Orthopedic Institute, Tianjin 300050, China; Tianjin Key Laboratory of Orthopedic Biomechanics and Medical Engineering, Tianjin 300050, China
| | - Yan Li
- Tianjin Hospital of Tianjin University (Tianjin Hospital), Tianjin 300211, China; Tianjin Orthopedic Institute, Tianjin 300050, China; Tianjin Key Laboratory of Orthopedic Biomechanics and Medical Engineering, Tianjin 300050, China
| | - Xinlong Ma
- Tianjin Hospital of Tianjin University (Tianjin Hospital), Tianjin 300211, China; Tianjin Orthopedic Institute, Tianjin 300050, China; Tianjin Key Laboratory of Orthopedic Biomechanics and Medical Engineering, Tianjin 300050, China
| | - Jianxiong Ma
- Tianjin Hospital of Tianjin University (Tianjin Hospital), Tianjin 300211, China; Tianjin Orthopedic Institute, Tianjin 300050, China; Tianjin Key Laboratory of Orthopedic Biomechanics and Medical Engineering, Tianjin 300050, China.
| |
Collapse
|
14
|
Rosenblatt KP, Zhang Z, Doss R, Gurnani PP, Grobman WA, Silver RM, Parry S, Reddy UM, Cao S, Haas DM. A multisite study to develop and validate first trimester, circulating microparticle biomarkers for tiered risk stratification of spontaneous preterm birth in nulliparas. Am J Obstet Gynecol 2025; 232:319.e1-319.e21. [PMID: 38789072 PMCID: PMC11584339 DOI: 10.1016/j.ajog.2024.05.032] [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: 01/12/2024] [Revised: 04/19/2024] [Accepted: 05/17/2024] [Indexed: 05/26/2024]
Abstract
BACKGROUND Despite much research, advances in early prediction of spontaneous preterm birth (sPTB) has been slow. The evolving field of circulating microparticle (CMP) biology may identify novel blood-based, and clinically useful, biomarkers. OBJECTIVE To test the ability of a previously identified, 7-marker set of CMP-derived proteins from the first trimester of pregnancy, in the form of an in vitro diagnostic multivariate index assay (IVDMIA), to stratify pregnant patients according to their risk for sPTB. STUDY DESIGN We employed a previously validated set of CMP protein biomarkers, utilizing mass spectrometry assays and a nested case-control design in a subset of participants from the Nulliparous Pregnancy Outcomes Study: monitoring mothers-to-be (nuMoM2b). We evaluated these biomarkers in the form of an IVDMIA to predict risk for sPTB at different gestational ages. Plasma samples collected at 9- to 13-weeks' gestation were analyzed. The IVDMIA assigned subjects to 1 of 3 sPTB risk categories: low risk (LR), moderate risk (MR), or high risk (HR). Independent validation on a set-aside set confirmed the IVDMIA's performance in risk stratification. RESULTS Samples from 400 participants from the nuMoM2b cohort were used for the study; of these, 160 delivered<37 weeks and 240 delivered at term. Through Monte Carlo simulation in which the validation results were adjusted based on actual weekly sPTB incidence rates in the nuMoM2b cohort, the IVDMIA stratifications demonstrated statistically significant differences among the risk groups in time-to-event (birth) analysis (P<.0001). The incidence-rate adjusted cumulative risks of sPTB at ≤32 weeks' gestation were 0.4%, 1.6%, and 7.5%, respectively for the LR, MR, and HR groups, respectively. Compared to the LR group, the corresponding risk ratios of the IVDMIA assigned MR and HR group were 4.25 (95% confidence interval [CI] 2.2-7.9) and 19.92 (95% CI 10.4-37.4), respectively. CONCLUSION A first trimester CMP protein biomarker panel can be used to stratify risk for sPTB at different gestational ages. Such a multitiered stratification tool could be used to assess risk early in pregnancy to enable timely clinical management and interventions, and, ultimately, to enable the development of tailored care pathways for sPTB prevention.
Collapse
Affiliation(s)
| | - Zhen Zhang
- Departments of Pathology and Oncology, Center for Biomarker Discovery and Translation, Johns Hopkins University School of Medicine, Baltimore, MD
| | | | | | - William A Grobman
- Department of Obstetrics and Gynecology, The Ohio State University School of Medicine, Columbus, OH
| | - Robert M Silver
- Department of Obstetrics and Gynecology, University of Utah School of Medicine, Salt Lake City, UT
| | - Samuel Parry
- Department of Obstetrics and Gynecology, University of Pennsylvania School of Medicine, Philadelphia, PA
| | - Uma M Reddy
- Department of Obstetrics and Gynecology, Columbia University School of Medicine, New York, NY
| | - Sha Cao
- Department of Biostatistics, Indiana University School of Medicine, Indianapolis, IN
| | - David M Haas
- Department of Obstetrics and Gynecology, Indiana University School of Medicine, Indianapolis, IN.
| |
Collapse
|
15
|
Huang HY, Zheng XN, Tian L. Vascular-Associated Mononuclear Phagocytes: First-Line Soldiers Ambushing Metastasis. Bioessays 2025; 47:e202400261. [PMID: 39988942 DOI: 10.1002/bies.202400261] [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: 11/05/2024] [Revised: 12/10/2024] [Accepted: 12/12/2024] [Indexed: 02/25/2025]
Abstract
Mononuclear phagocytes (MPs), which consist of dendritic cells, monocytes, and macrophages, are distributed throughout the body and actively eliminate invading microorganisms and abnormal cells. Depending on the local microenvironment, MPs manifest considerably various lifespans and phenotypes to maintain tissue homeostasis. Vascular-associated mononuclear phagocytes (VaMPs) are the special subsets of MPs that are localized either within the lumen side or on the apical surface of vessels, acting as the critical sentinels to recognize and defend against disseminated tumor cells. In this review, we introduce three major types of VaMPs, patrolling monocytes, Kupffer cells, and perivascular macrophages, and discuss their emerging roles in immunosurveillance during incipient metastasis. We also explore the roles of lineage-determining transcription factors and cell surface receptors that endow VaMPs with potent anti-tumor activity. Finally, we highlight the molecular and cellular mechanisms that drive the phenotypic plasticity of VaMPs and summarize combinatory strategies for targeting VaMPs in overt metastasis.
Collapse
Affiliation(s)
- Han-Ying Huang
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
| | - Xin-Nan Zheng
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
| | - Lin Tian
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
| |
Collapse
|
16
|
Zhu Y, Gao L, Zhang J, Li M, Zhou J, Zhou J. Extracellular vesicle-packaged PKM2 from endometriotic stromal cells promotes endometrial collagen I deposition by inhibiting autophagy in endometriosis. Cell Signal 2025; 127:111523. [PMID: 39586523 DOI: 10.1016/j.cellsig.2024.111523] [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: 07/16/2024] [Revised: 11/04/2024] [Accepted: 11/20/2024] [Indexed: 11/27/2024]
Abstract
Aberrant endometrial collagen I deposition during the implantation window impairs endometrial stromal cell (ESC) decidualization, which may contribute to lower pregnancy rate in endometriosis (EMs) patients with in vitro fertilization (IVF) treatment. However, the underlying mechanism of eutopic aberrant endometrium collagen I deposition in EMs remains unclear. In this study, we found increased endometrial collagen I and defective decidualization in the mid-secretory phase of EMs patients, while the level of eutopic ESCs' autophagy was decreased, which was an important mechanism of intracellular collagen degradation. Lower ESCs' autophagy level may cause the endometrial collagen I deposition in EMs. Furthermore, in vivo and in vitro studies showed that the extracellular vesicles derived from the ectopic ESCs of EMs patients (EMs-EVs) encapsulated higher PKM2 inhibited autophagy of the ESCs accompanied by an increase of collagen I. We also found that the constructed EMs-EVsAd-PKM2 with PKM2 overexpression inhibited ESCs' autophagy by activating the Akt/mTOR signaling pathway. And the expressions of PKM2, p-Akt and p-mTOR were also increased in the endometrium of EMs patients. Collectively, these data showed that EMs-EVs delivering PKM2 inhibited autophagy inducing aberrant endometrial collagen I deposition via the Akt/mTOR signaling pathway to impair decidualization, which provided a potential therapeutic target for improving the IVF pregnancy rate in EMs patients.
Collapse
Affiliation(s)
- Yuan Zhu
- Center for Reproductive Medicine and Obstetrics and Gynecology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Liang Gao
- Center for Reproductive Medicine and Obstetrics and Gynecology, Drum Tower Clinic Medical College of Nanjing Medical University, Nanjing, China
| | - Jingyu Zhang
- Center for Reproductive Medicine and Obstetrics and Gynecology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Mengyun Li
- Center for Reproductive Medicine and Obstetrics and Gynecology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Jidong Zhou
- Center for Reproductive Medicine and Obstetrics and Gynecology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Jianjun Zhou
- Center for Reproductive Medicine and Obstetrics and Gynecology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
| |
Collapse
|
17
|
Zhang Q, Liu X, Wei Q, Xiong S, Luo W, Zhou Y, Cao J, Xu X, Liu R, Tang X, Zhang W, Luo B. Apoptotic breast cancer cells after chemotherapy induce pro-tumour extracellular vesicles via LAP-competent macrophages. Redox Biol 2025; 80:103485. [PMID: 39756316 PMCID: PMC11758215 DOI: 10.1016/j.redox.2024.103485] [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: 12/04/2024] [Accepted: 12/27/2024] [Indexed: 01/07/2025] Open
Abstract
Chemotherapy is important in the systemic therapy for breast cancer. However, after chemotherapy, the left living tumour cells are more progressive. There is an urgent need to study the underlying mechanism which is still unclear to further improve the therapeutic efficacy of chemotherapy in breast cancer. Here we find a pro-tumour effect of the apoptotic cells induced by the chemotherapy, which is mediated by a new subset of macrophages undergoing LC3-associated phagocytosis (LAP). By transferring exosomal S100A11 into the living tumour cells after chemotherapy, the macrophage exhibits a more pro-tumour phenotype than classic M2-type macrophages. Moreover, S100A11 binds to IFITM3, inducing Akt phosphorylation of living tumour cells after chemotherapy, which promotes tumour progression. Of note, Akt inhibitor can enhance the therapeutic effcicay of chemotherapy in breast cancer. This study provides a novel mechanistic link between tumour-associated macrophages and breast cancer, uncovering Akt as a potential therapeutic target to improve chemotherapy efficacy.
Collapse
Affiliation(s)
- Qi Zhang
- Department of Ultrasound, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China; Guangdong Provincial Key Laboratory of Malignant Tumour Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China; Department of Ultrasound, The First Affiliated Hospital with Nanjing Medical University, Nanjing, 210029, China
| | - Xiaodi Liu
- Department of Ultrasound, Laboratory of Ultrasound Imaging and Drug, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Qiuxia Wei
- Department of Ultrasound, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China; Guangdong Provincial Key Laboratory of Malignant Tumour Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Shiyu Xiong
- Department of Ultrasound, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China; Guangdong Provincial Key Laboratory of Malignant Tumour Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Wanrong Luo
- Department of Ultrasound, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China; Guangdong Provincial Key Laboratory of Malignant Tumour Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Yingshi Zhou
- Department of Ultrasound, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China; Guangdong Provincial Key Laboratory of Malignant Tumour Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Jincheng Cao
- Department of Ultrasound, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China; Guangdong Provincial Key Laboratory of Malignant Tumour Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Xiaolin Xu
- Department of Ultrasound, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Rongbin Liu
- Department of Ultrasound, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Xinyu Tang
- Department of Breast Surgery, Department of General Surgery, The First Affiliated Hospital with Nanjing Medical University, Nanjing, 210029, China; Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Jiangsu Collaborative Innovation Center For Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, 211166, China.
| | - Wenyue Zhang
- Department of Ultrasound, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China; Guangdong Provincial Key Laboratory of Malignant Tumour Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China.
| | - Baoming Luo
- Department of Ultrasound, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China.
| |
Collapse
|
18
|
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
|
19
|
Wang J, Zhu Y, He Y, Shao W. TAM-derived exosomal miR-589-3p accelerates ovarian cancer progression through BCL2L13. J Ovarian Res 2025; 18:36. [PMID: 39985077 PMCID: PMC11846191 DOI: 10.1186/s13048-025-01618-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 02/05/2025] [Indexed: 02/24/2025] Open
Abstract
BACKGROUND Tumor-associated macrophages (TAM) are critical elements of intercellular communication in tumor microenvironment (TME), and exosomes are key mediators between tumor cells and the TME. According to previous reports, miRNAs exert a pivotal role in ovarian cancer (OC) development. The purpose of this work was to explore the function of TAM-derived exosomal miR-589-3p in OC development and elucidate the underlying molecular mechanisms. METHODS First, peripheral blood mononuclear cells (PBMC) were treated with IL-4 and IL-13 to polarize them into M2-type macrophages. Exosomes were separated from M2-type macrophages, and the physical properties of exosomes were evaluated using transmission electron microscopy (TEM) and nanoparticle tracking analysis (NTA). Next, quantitative reverse-transcriptase polymerase chain reaction (qRT-PCR) was applied to examine the expression of relevant genes. Subsequently, Targetscan and miRDB were utilized to predict miR-589-3p target genes, and then the interaction between miR-589-3p and BCL2L13 was verified by dual luciferase assay and RNA Binding Protein Immunoprecipitation (RIP) assay. Finally, Cell Counting Kit-8 (CCK-8) and flow cytometry experiments were employed to explore the changes in the proliferative and apoptotic abilities of OC cells. RESULTS In this research, we demonstrated that TAM-derived exosomes facilitated OC cell proliferation and suppressed OC cell apoptosis. Then, qRT-PCR results indicated that miR-589-3p were markedly elevated after co-culture of TAM-derived exosomes with OC cells. In addition, we discovered that miR-589-3p was bound to BCL-2-like protein 13 (BCL2L13), which was confirmed through luciferase assay and RIP assay. Furthermore, functional analysis displayed that TAM-derived exosomes treated with miR-589-3p inhibitor attenuated the promotion of OC cell progression by exosomes. CONCLUSION TAM-derived exosomal miR-589-3p enhanced OC progression through BCL2L13, which offers a novel for OC therapy.
Collapse
Affiliation(s)
- Jianqing Wang
- Department of Gynecology and Obstetrics, Yancheng First People's Hospital, Yancheng Clinical College of Xuzhou Medical University, Yancheng, Jiangsu, 224002, China
| | - Yan Zhu
- Department of Gynecology and Obstetrics, Yancheng First People's Hospital, Yancheng Clinical College of Xuzhou Medical University, Yancheng, Jiangsu, 224002, China
| | - Yang He
- Department of Gynecology and Obstetrics, Yancheng First People's Hospital, Yancheng Clinical College of Xuzhou Medical University, Yancheng, Jiangsu, 224002, China
| | - Weiwei Shao
- Department of Pathology, Yancheng First People's Hospital, Yancheng Clinical College of Xuzhou Medical University, Yancheng, Jiangsu, 224002, China.
- Department of Pathology, Yancheng Clinical College of Xuzhou Medical University, Yancheng First People's Hospital, No. 166, Yulong West Road, Yancheng, Jiangsu, 224002, China.
| |
Collapse
|
20
|
Liu X, Liu J, Wang X, Zou Y, Tao X, Li J, Ye M, Xu W, Deng Y, Liu L, Sun J, Zhang Q. Cancer-secreted exosomal miR-1246 promotes colorectal cancer liver metastasis by activating hepatic stellate cells. Mol Med 2025; 31:68. [PMID: 39979806 PMCID: PMC11841005 DOI: 10.1186/s10020-025-01112-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 01/31/2025] [Indexed: 02/22/2025] Open
Abstract
Colorectal cancer liver metastases (CRLM) are the primary cause of mortality in colorectal cancer (CRC) patients, highlighting the importance of understanding the underlying mechanisms. The tumor microenvironment (TME) and its interaction with tumor cells play a crucial role in CRLM progression. Notably, the stability and peak levels of tumor-derived exosomal miRNAs facilitate intercellular communication in the TME. Hepatic stellate cells (HSCs), key liver mesenchymal cells, constitute about 33% of the liver's nonsolid cell population and exhibit plasticity. However, the specific role of tumor-derived exosomal miRNAs in the crosstalk between HSCs and tumor cells during the CRLM process remains unclear. We studied CRC-secreted exosomal miR-1246 and its impact on HSCs, as well as its effects on CRC cell proliferation and metastasis. Our findings demonstrate that CRC-secreted exosomal miR-1246 can be internalized by HSCs, leading to their activation and facilitating the metastatic potential of CRC cells. Mechanistically, exosomal miR-1246 targets INSIG1, resulting in SREBP2 nucleation and cholesterol metabolism alterations. This accumulation of free cholesterol (FC) regulates the TLR4/NF-κB/TGF-β pathway, promoting HSC activation. Activated HSCs, in turn, enhance liver metastasis of CRC cells through the TNFSF13/TNFRSF13B axis. Our study reveals the role of CRC-secreted exosomal miR-1246 in triggering HSC activation and reprogramming the TME, ultimately facilitating liver metastasis in CRC patients. Exosomal miR-1246 could serve as a potential non-invasive biomarker for predicting colorectal cancer liver metastasis, enhancing our understanding of CRC-associated liver metastases.
Collapse
Affiliation(s)
- Xiaolong Liu
- Department of General Surgery, The Third Affiliated Hospital of Southern Medical University, 183 West Zhongshan Avenue, Guangzhou, Guangdong, 510630, People's Republic of China
| | - Jialong Liu
- Department of General Surgery, The Third Affiliated Hospital of Southern Medical University, 183 West Zhongshan Avenue, Guangzhou, Guangdong, 510630, People's Republic of China
| | - Xuanyin Wang
- Department of General Surgery, The Third Affiliated Hospital of Southern Medical University, 183 West Zhongshan Avenue, Guangzhou, Guangdong, 510630, People's Republic of China
| | - Yang Zou
- Department of General Surgery, The Third Affiliated Hospital of Southern Medical University, 183 West Zhongshan Avenue, Guangzhou, Guangdong, 510630, People's Republic of China
| | - Xinyi Tao
- Department of General Surgery, The Third Affiliated Hospital of Southern Medical University, 183 West Zhongshan Avenue, Guangzhou, Guangdong, 510630, People's Republic of China
| | - Jingyu Li
- Department of Pathology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, No.106, Zhongshan 2 Road, Guangzhou, Guangdong, 510080, People's Republic of China
| | - Mengnan Ye
- Department of Pathology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, No.106, Zhongshan 2 Road, Guangzhou, Guangdong, 510080, People's Republic of China
| | - Wanbei Xu
- Department of General Surgery, The Third Affiliated Hospital of Southern Medical University, 183 West Zhongshan Avenue, Guangzhou, Guangdong, 510630, People's Republic of China
| | - Yunyao Deng
- Department of General Surgery, The Third Affiliated Hospital of Southern Medical University, 183 West Zhongshan Avenue, Guangzhou, Guangdong, 510630, People's Republic of China
| | - Lixin Liu
- Department of General Surgery, The Third Affiliated Hospital of Southern Medical University, 183 West Zhongshan Avenue, Guangzhou, Guangdong, 510630, People's Republic of China.
| | - Jingbo Sun
- Department of General Surgery, The Third Affiliated Hospital of Southern Medical University, 183 West Zhongshan Avenue, Guangzhou, Guangdong, 510630, People's Republic of China.
| | - Qingling Zhang
- Department of Pathology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, No.106, Zhongshan 2 Road, Guangzhou, Guangdong, 510080, People's Republic of China.
| |
Collapse
|
21
|
Wang Z, Ren Y, Li Y, Zhang Y, Bai S, Hou W, Zhang W, Yao Y, Zhao H, Wang M, Luo Y, Pang G, Du J. MiR-186-5p carried by M2 macrophage-derived exosomes downregulates TRPP2 expression in airway smooth muscle to alleviate asthma progression. Int Immunopharmacol 2025; 148:114107. [PMID: 39884080 DOI: 10.1016/j.intimp.2025.114107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 01/15/2025] [Accepted: 01/15/2025] [Indexed: 02/01/2025]
Abstract
Bronchial asthma (asthma) is a chronic inflammatory disease of the airways that remains an unresolved problem. Reportedly M2 macrophages and exosomes play a role in inflammation, including asthma. We investigated the roles of M2 macrophage-derived exosomes (M2-Exos) effect in asthmatic progression by using ovalbumin (OVA) induced asthmatic mice model. M2-Exos significantly ameliorated the pulmonary inflammatory response and airway hyperresponsiveness in asthmatic mice and suppressed aberrant proliferation and transient receptor potential polycystic protein 2(TRPP2) expression in LPS-stimulated primary airway smooth muscle cells (ASMCs). Then, we found that miR-186-5p of M2-Exos could target TRPP2 through online database analysis. However, miR-186-5p downregulation by miR-186-5p inhibitors decreased the protective effect of M2-Exos in asthmatic mouse and cellular models. miR-186-5p was identified and selectively combined with the polycystin-2 gene encoding TRPP2 protein, inhibited TRPP2 protein production, and downregulated TRPP2 expression. A reduction in the number of TRPP2 calcium (Ca) channels formed on the cell membrane leads to a decreased intracellular Ca2+ concentration ([Ca2+] i), causing reduced ASMC contraction and proliferation, thereby improving airway hyperresponsiveness and airway remodeling in asthma. Collectively, we conclude that M2 exosomal miR-186-5p to alleviate asthma progression and airway hyperresponsiveness though downregulating TRPP2 expression. These results may offer a novel insight to the treatment and drug delivery of asthma.
Collapse
Affiliation(s)
- Zunyun Wang
- Ciechanover Institute of Precision and Regenerative Medicine, School of Medicine, The Chinese University of Hong Kong Shenzhen Guangdong China
| | - Yan Ren
- School of Basic Medical Sciences Anhui Medical University Hefei Anhui China; The Second Affiliated Hospital, School of Medicine, The Chinese University of Hong Kong Shenzhen & Longgang District People's Hospital of Shenzhen Shenzhen Guangdong China
| | - Yicong Li
- School of Basic Medical Sciences Anhui Medical University Hefei Anhui China; The Second Affiliated Hospital, School of Medicine, The Chinese University of Hong Kong Shenzhen & Longgang District People's Hospital of Shenzhen Shenzhen Guangdong China
| | - Yuxin Zhang
- School of Basic Medical Sciences Anhui Medical University Hefei Anhui China; The Second Affiliated Hospital, School of Medicine, The Chinese University of Hong Kong Shenzhen & Longgang District People's Hospital of Shenzhen Shenzhen Guangdong China
| | - Suwen Bai
- The Second Affiliated Hospital, School of Medicine, The Chinese University of Hong Kong Shenzhen & Longgang District People's Hospital of Shenzhen Shenzhen Guangdong China
| | - Wenxuan Hou
- School of Basic Medical Sciences Anhui Medical University Hefei Anhui China; The Second Affiliated Hospital, School of Medicine, The Chinese University of Hong Kong Shenzhen & Longgang District People's Hospital of Shenzhen Shenzhen Guangdong China
| | - Wenjun Zhang
- School of Basic Medical Sciences Anhui Medical University Hefei Anhui China; The Second Affiliated Hospital, School of Medicine, The Chinese University of Hong Kong Shenzhen & Longgang District People's Hospital of Shenzhen Shenzhen Guangdong China
| | - Yanheng Yao
- School of Basic Medical Sciences Anhui Medical University Hefei Anhui China; The Second Affiliated Hospital, School of Medicine, The Chinese University of Hong Kong Shenzhen & Longgang District People's Hospital of Shenzhen Shenzhen Guangdong China
| | - Hongxian Zhao
- Ciechanover Institute of Precision and Regenerative Medicine, School of Medicine, The Chinese University of Hong Kong Shenzhen Guangdong China
| | - Minghua Wang
- The Second Affiliated Hospital, School of Medicine, The Chinese University of Hong Kong Shenzhen & Longgang District People's Hospital of Shenzhen Shenzhen Guangdong China
| | - Yumei Luo
- The Second Affiliated Hospital, School of Medicine, The Chinese University of Hong Kong Shenzhen & Longgang District People's Hospital of Shenzhen Shenzhen Guangdong China
| | - Gang Pang
- School of Basic Medical Sciences Anhui Medical University Hefei Anhui China.
| | - Juan Du
- Ciechanover Institute of Precision and Regenerative Medicine, School of Medicine, The Chinese University of Hong Kong Shenzhen Guangdong China; The Second Affiliated Hospital, School of Medicine, The Chinese University of Hong Kong Shenzhen & Longgang District People's Hospital of Shenzhen Shenzhen Guangdong China.
| |
Collapse
|
22
|
Shinde U, Balasinor NH, Ravichandran V, Kumar AS, Gunasekaran VP. "Extracellular Vesicle DNA: Advances and Applications as a Non-Invasive Biomarker in Disease Diagnosis and Treatment". Clin Chim Acta 2025; 568:120125. [PMID: 39793847 DOI: 10.1016/j.cca.2025.120125] [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: 11/19/2024] [Revised: 01/06/2025] [Accepted: 01/06/2025] [Indexed: 01/13/2025]
Abstract
Extracellular vesicles (EVs) are nanoscale, membrane-enclosed structures released by cells into the extracellular milieu. These vesicles encapsulate a diverse array of molecular constituents, including nucleic acids, proteins, and lipids, which provide insights into the physiological or pathological conditions of their parent cells. Despite their potential, the study of EV-derived DNA (EV-DNA) has gathered relatively limited attention. This review aims to present a thorough examination of the emerging knowledge surrounding the utility of EV-DNA as a non-invasive biomarker across a spectrum of diseases. The review delves into various mechanisms underlying DNA packaging within EVs and the prevalent methodologies employed for extraction of EV-DNA. The relevance of EV-DNA is assessed across numerous health conditions, notably cancer, cardiovascular diseases, neurodegenerative disorders, infectious diseases, and pregnancy-related complications. The use of EV-DNA for cancer mutation detection has demonstrated remarkable sensitivity and specificity, thereby enhancing both diagnostic accuracy and therapeutic monitoring. In the context of cardiovascular diseases, EV-DNA serves as a predictive marker for events such as myocardial infarctions and shows a correlation with the severity of the disease. With respect to neurodegenerative conditions, including Parkinson's and Alzheimer's, EV-DNA contributes to the understanding of disease mechanisms and progression. Additionally, it plays an essential role in modulating immune tolerance and facilitating communication between maternal and fetal systems. Although there is a pressing need for standardized protocols for EV isolation and DNA analysis to facilitate clinical implementation, the prospect of EV-DNA as a non-invasive biomarker for diagnostic and prognostic purposes across diverse pathological conditions is considerable.
Collapse
Affiliation(s)
- Uma Shinde
- Center for Drug Discovery and Development, Amity Institute of Biotechnology, Amity University Mumbai (AUM), Maharashtra, India
| | - Nafisa Huseni Balasinor
- ICMR-National Institute for Research in Reproductive and Child Health (ICMR- NIRRCH), Parel, Mumbai, India
| | - Vinothkannan Ravichandran
- Center for Drug Discovery and Development, Amity Institute of Biotechnology, Amity University Mumbai (AUM), Maharashtra, India
| | - Aw Santhosh Kumar
- Center for Drug Discovery and Development, Amity Institute of Biotechnology, Amity University Mumbai (AUM), Maharashtra, India; California University of Science & Medicine, CA, United States of America
| | - Vinoth Prasanna Gunasekaran
- Center for Drug Discovery and Development, Amity Institute of Biotechnology, Amity University Mumbai (AUM), Maharashtra, India.
| |
Collapse
|
23
|
Kuang L, Wu L, Li Y. Extracellular vesicles in tumor immunity: mechanisms and novel insights. Mol Cancer 2025; 24:45. [PMID: 39953480 PMCID: PMC11829561 DOI: 10.1186/s12943-025-02233-w] [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: 09/30/2024] [Accepted: 01/14/2025] [Indexed: 02/17/2025] Open
Abstract
Extracellular vesicles (EVs), nanoscale vesicles secreted by cells, have attracted considerable attention in recent years due to their role in tumor immunomodulation. These vesicles facilitate intercellular communication by transporting proteins, nucleic acids, and other biologically active substances, and they exhibit a dual role in tumor development and immune evasion mechanisms. Specifically, EVs can assist tumor cells in evading immune surveillance and attack by impairing immune cell function or modulating immunosuppressive pathways, thereby promoting tumor progression and metastasis. Conversely, they can also transport and release immunomodulatory factors that stimulate the activation and regulation of the immune system, enhancing the body's capacity to combat malignant diseases. This dual functionality of EVs presents promising avenues and targets for tumor immunotherapy. By examining the biological characteristics of EVs and their influence on tumor immunity, novel therapeutic strategies can be developed to improve the efficacy and relevance of cancer treatment. This review delineates the complex role of EVs in tumor immunomodulation and explores their potential implications for cancer therapeutic approaches, aiming to establish a theoretical foundation and provide practical insights for the advancement of future EVs-based cancer immunotherapy strategies.
Collapse
Affiliation(s)
- Liwen Kuang
- School of Medicine, Chongqing University, Chongqing, China
| | - Lei Wu
- Department of Medical Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Yongsheng Li
- School of Medicine, Chongqing University, Chongqing, China.
- Department of Medical Oncology, Chongqing University Cancer Hospital, Chongqing, China.
| |
Collapse
|
24
|
Vakili-Ghartavol Z, Deli H, Shadboorestan A, Sahebnasagh R, Motevaseli E, Ghahremani MH. Exosomes and their distinct integrins transfer the characteristics of oxaliplatin- and 5-FU-resistant behaviors in colorectal cancer cells. Mol Med 2025; 31:49. [PMID: 39915745 PMCID: PMC11803997 DOI: 10.1186/s10020-025-01110-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: 10/11/2024] [Accepted: 01/29/2025] [Indexed: 02/09/2025] Open
Abstract
BACKGROUND Exosomes are communication carriers and suitable biomarker candidates due to their cargoes with specific dynamic profiles. Integrins, as valuable prognostic markers in cancer, have importance in exosome-cell interaction. However, the role of exosome integrins in chemoresistant colorectal cancer remained unclear. METHODS Oxaliplatin- and 5-FU-resistant cells (OXR and FUR) were established from human HCT-116 cells of colorectal cancer. Exosomes were collected from untreated and treated cells with oxaliplatin or 5-FU. Exosomes were isolated via ultracentrifugation and characterized using DLS and electron microscopy to evaluate size and morphology. Western blot analysis was employed to identify exosomal markers. The effects of exosomes on parental cells were examined using various methods, including MTT assay for proliferation, wound healing assay for migration, flow cytometry for cell cycle and apoptosis analysis, Matrigel-coated transwell inserts for invasion, and western blot for integrin expression evaluation. RESULTS Exosome integrins determine resistance behaviors in cells. We observed that exosomes from OXR cells or OXR cells treated with oxaliplatin increased ITGβ3 expression and decreased ITGβ4 expression in parental cells, resulting in distinct resistance behaviors. Exosomes from FUR cells or FUR cells treated with 5-FU reduced ITGβ4 levels and elevated ITGαv levels in parental cells, leading to varying degrees of invasive resistance behaviors. These findings suggest that exosome integrins may affect these behaviors. High ITGβ3 exosomes induced oxaliplatin resistance behaviors in parental cells. Lowering ITGβ3 levels in these exosomes inhibited the resistance behaviors observed in these cells. FUR exosomes that overexpressed ITGαv or ITGβ4 resulted in invasive 5-FU resistance behaviors in parental cells. A reduction in these exosome integrin levels led to moderate invasive behaviors. The decrease of ITGβ4 in FUR cell exosomes inhibited resistant migration and proliferation in parental cells. A twofold reduction of ITGαv in FUR cell exosomes resulted in a threefold decrease in invasion and inhibited migration in parental cells compared to those treated with high ITGαv exosomes. CONCLUSION Our findings reveal that, despite discrepancies between cellular integrin patterns and cellular behaviors, the levels of exosomal ITGβ3, ITGαv, or ITGβ4 could serve as potential diagnostic and therapeutic markers for resistance to oxaliplatin and 5-FU in future cancer treatments.
Collapse
Affiliation(s)
- Zeynab Vakili-Ghartavol
- Department of Molecular Medicine, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Hoda Deli
- Department of Molecular Medicine, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Amir Shadboorestan
- Department of Toxicology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Roxana Sahebnasagh
- Department of Molecular Medicine, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Elahe Motevaseli
- Department of Molecular Medicine, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Hossein Ghahremani
- Department of Molecular Medicine, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
25
|
Saadh MJ, Allela OQB, Kareem RA, Ballal S, Chahar M, Saini S, Prasad GVS, Sameer HN, Hamad AK, Athab ZH, Adil M. The role of exosomal non-coding RNAs in the breast cancer tumor microenvironment. Funct Integr Genomics 2025; 25:32. [PMID: 39891771 DOI: 10.1007/s10142-025-01531-2] [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: 10/07/2024] [Revised: 01/06/2025] [Accepted: 01/08/2025] [Indexed: 02/03/2025]
Abstract
The leading form of cancer affecting females globally is breast cancer, characterized by an unregulated growth of cells within the breast. Therefore, examining breast tissue is crucial in accurately identifying and treating this disease. Exosomes are very small enclosures bounded by a layer of cells and produced by a variety of cells present in the cancerous tissue surroundings. They play a crucial role in several biological functions in cancerous tumors. These exosomes carry non-coding RNAs (ncRNAs) and are discharged into the TME, where they are instrumental in the development and advancement of tumors. Additionally, the ncRNAs enclosed in exosomes act as significant mediators of communication within cells. Consequently, there is limited comprehension regarding the precise roles and targets of exosomal RNA in regulation, as research in this area is still in its preliminary phases. This piece provides a comprehensive overview of the latest studies on exosomes, delving into their impact on the behavior of cancer cells and immune cells. Moreover, it presents a compilation of the diverse forms of non-coding RNA molecules found in exosomes released by both cancerous and supportive cells, including circular RNAs, microRNAs, and long non-coding RNAs. Current research has proven the noteworthy influence that non-coding RNA molecules have on the progression, proliferation, drug resistance, and immune responses of breast cancer cells.
Collapse
Affiliation(s)
- Mohamed J Saadh
- Faculty of Pharmacy, Middle East University, 11831, Amman, Jordan
| | | | | | - Suhas Ballal
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Mamata Chahar
- Department of Chemistry, NIMS Institute of Engineering & Technology, NIMS University Rajasthan, Jaipur, India
| | - Suman Saini
- Department of Applied Sciences, Chandigarh Engineering College, Chandigarh Group of Colleges-Jhanjeri, 140307, Mohali, Punjab, India
| | - G V Siva Prasad
- Department of Basic Sciences and Humanities, Raghu Engineering College, Visakhapatnam, Andhra Pradesh, 531162, India
| | - Hayder Naji Sameer
- Collage of Pharmacy, National University of Science and Technology, 64001, Dhi Qar, Iraq
| | | | - Zainab H Athab
- Department of Pharmacy, Al-Zahrawi University College, Karbala, Iraq
| | - Mohaned Adil
- Pharmacy college, Al-Farahidi University, 00964, Baghdad, Iraq
| |
Collapse
|
26
|
Wang L, Zhou S, Ruan Y, Wu X, Zhang X, Li Y, Ying D, Lu Y, Tian Y, Cheng G, Zhang J, Lv K, Zhou X. Hypoxia-Challenged Pancreatic Adenocarcinoma Cell-Derived Exosomal circR3HCC1L Drives Tumor Growth Via Upregulating PKM2 Through Sequestering miR-873-5p. Mol Biotechnol 2025; 67:762-777. [PMID: 38526683 DOI: 10.1007/s12033-024-01091-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Accepted: 01/24/2024] [Indexed: 03/27/2024]
Abstract
Pancreatic adenocarcinoma (PAAD) is a fatal disease with poor survival. Increasing evidence show that hypoxia-induced exosomes are associated with cancer progression. Here, we aimed to investigate the function of hsa_circ_0007678 (circR3HCC1L) and hypoxic PAAD cell-derived exosomal circR3HCC1L in PAAD progression. Through the exoRBase 2.0 database, we screened for a circular RNA circR3HCC1L related to PAAD. Changes of circR3HCC1L in PAAD samples and cells were analyzed with real-time quantitative polymerase chain reaction (RT-qPCR). Cell proliferation, migration, invasion were analyzed by colony formation, cell counting, and transwell assays. Measurements of glucose uptake and lactate production were done using corresponding kits. Several protein levels were detected by western blotting. The regulation mechanism of circR3HCC1L was verified by dual-luciferase reporter, RNA immunoprecipitation, and RNA pull-down assays. Exosomes were separated by differential ultracentrifugation. Animal experiments were used to verify the function of hypoxia-derived exosomal circR3HCC1L. CircR3HCC1L was upregulated in PAAD samples and hypoxic PAAD cells. Knockdown of circR3HCC1L decreased hypoxia-driven PAAD cell proliferation, migration, invasion, and glycolysis. Hypoxic PAAD cell-derived exosomes had higher levels of circR3HCC1L, hypoxic PAAD cell-derived exosomal circR3HCC1L promoted normoxic cancer cell malignant transformation and glycolysis in vitro and xenograft tumor growth in mouse models in vivo. Mechanistically, circR3HCC1L regulated pyruvate kinase M2 (PKM2) expression via sponging miR-873-5p. Also, PKM2 overexpression or miR-873-5p silencing offset circR3HCC1L knockdown-mediated effects on hypoxia-challenged PAAD cell malignant transformation and glycolysis. Hypoxic PAAD cell-derived exosomal circR3HCC1L facilitated PAAD progression through the miR-873-5p/PKM2 axis, highlighting the contribution of hypoxic PAAD cell-derived exosomal circR3HCC1L in PAAD.
Collapse
Affiliation(s)
- Luoluo Wang
- Department of Abdominal Minimally Invasive Surgery, Ningbo Medical Center Lihuili Hospital, Ningbo University, No.1111, Jiangnan Road, Yinzhou District, Ningbo, 315040, Zhejiang, China
| | - Shuping Zhou
- Ningbo College of Health Sciences, No.51, Xuefu Road, Yinzhou District, Ningbo, 315040, Zhejiang, China.
| | - Yi Ruan
- Department of Abdominal Minimally Invasive Surgery, Ningbo Medical Center Lihuili Hospital, Ningbo University, No.1111, Jiangnan Road, Yinzhou District, Ningbo, 315040, Zhejiang, China
| | - Xiang Wu
- Department of Abdominal Minimally Invasive Surgery, Ningbo Medical Center Lihuili Hospital, Ningbo University, No.1111, Jiangnan Road, Yinzhou District, Ningbo, 315040, Zhejiang, China
- Medical School of Ningbo University, Ningbo, 315040, Zhejiang, China
| | - Xueming Zhang
- Department of Abdominal Minimally Invasive Surgery, Ningbo Medical Center Lihuili Hospital, Ningbo University, No.1111, Jiangnan Road, Yinzhou District, Ningbo, 315040, Zhejiang, China
| | - Yi Li
- College of Computer Science and Artificial Intelligence Wenzhou University, Wenzhou, 325000, Zhejiang, China
| | - Dongjian Ying
- Department of Abdominal Minimally Invasive Surgery, Ningbo Medical Center Lihuili Hospital, Ningbo University, No.1111, Jiangnan Road, Yinzhou District, Ningbo, 315040, Zhejiang, China
| | - Yeting Lu
- Department of Abdominal Minimally Invasive Surgery, Ningbo Medical Center Lihuili Hospital, Ningbo University, No.1111, Jiangnan Road, Yinzhou District, Ningbo, 315040, Zhejiang, China
| | - Yuan Tian
- Department of Abdominal Minimally Invasive Surgery, Ningbo Medical Center Lihuili Hospital, Ningbo University, No.1111, Jiangnan Road, Yinzhou District, Ningbo, 315040, Zhejiang, China
| | - Gong Cheng
- Department of Abdominal Minimally Invasive Surgery, Ningbo Medical Center Lihuili Hospital, Ningbo University, No.1111, Jiangnan Road, Yinzhou District, Ningbo, 315040, Zhejiang, China
| | - Jing Zhang
- Department of Abdominal Minimally Invasive Surgery, Ningbo Medical Center Lihuili Hospital, Ningbo University, No.1111, Jiangnan Road, Yinzhou District, Ningbo, 315040, Zhejiang, China
| | - Kaiji Lv
- Department of Abdominal Minimally Invasive Surgery, Ningbo Medical Center Lihuili Hospital, Ningbo University, No.1111, Jiangnan Road, Yinzhou District, Ningbo, 315040, Zhejiang, China
| | - Xinhua Zhou
- Department of Abdominal Minimally Invasive Surgery, Ningbo Medical Center Lihuili Hospital, Ningbo University, No.1111, Jiangnan Road, Yinzhou District, Ningbo, 315040, Zhejiang, China.
| |
Collapse
|
27
|
Cesana B, Cochet C, Filhol O. New players in the landscape of renal cell carcinoma bone metastasis and therapeutic opportunities. Int J Cancer 2025; 156:475-487. [PMID: 39306698 PMCID: PMC11622000 DOI: 10.1002/ijc.35181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 08/09/2024] [Accepted: 08/20/2024] [Indexed: 12/07/2024]
Abstract
Approximately one-third of advanced renal cell carcinoma (RCC) patients develop osteolytic bone metastases, leading to skeletal complications. In this review, we first provide a comprehensive perspective of seminal studies on bone metastasis of RCC describing the main molecular modulators and growth factor signaling pathways most important for the RCC-stimulated osteoclast-mediated bone destruction. We next focus on newer developments revealing with in-depth details, the bidirectional interplay between renal cancer cells and the immune and stromal microenvironment that can through epigenetic reprogramming, profoundly affect the behaviors of transformed cells. Understanding their mechanistic interactions is of paramount importance for advancing both fundamental and translational research. These new investigations into the landscape of RCC-bone metastasis offer novel insights and identify potential avenues for future therapeutic interventions.
Collapse
Affiliation(s)
- Beatrice Cesana
- University Grenoble Alpes, INSERM, CEA, IRIG‐Biosanté, UMR 1292GrenobleFrance
| | - Claude Cochet
- University Grenoble Alpes, INSERM, CEA, IRIG‐Biosanté, UMR 1292GrenobleFrance
| | - Odile Filhol
- University Grenoble Alpes, INSERM, CEA, IRIG‐Biosanté, UMR 1292GrenobleFrance
| |
Collapse
|
28
|
Zhang Y, Wang X, Gu Y, Liu T, Zhao X, Cheng S, Duan L, Huang C, Wu S, Gao S. Complement C3 of tumor-derived extracellular vesicles promotes metastasis of RCC via recruitment of immunosuppressive myeloid cells. Proc Natl Acad Sci U S A 2025; 122:e2420005122. [PMID: 39847320 PMCID: PMC11789090 DOI: 10.1073/pnas.2420005122] [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: 09/30/2024] [Accepted: 12/13/2024] [Indexed: 01/24/2025] Open
Abstract
Heterogeneous roles of complement C3 have been implicated in tumor metastasis and are highly context dependent. However, the underlying mechanisms linking C3 to tumor metastasis remain elusive in renal cell carcinoma (RCC). Here, we demonstrate that C3 of RCC cell-derived extracellular vesicles (EVs) contributes to metastasis via polarizing tumor-associated macrophages (TAMs) into the immunosuppressive phenotype and recruiting polymorphonuclear myeloid-derived suppressor cells (PMN-MDSCs). Mechanistically, EV C3 induces the secretion of CCL2 and CXCL1 by lung macrophages and subsequently enhances TAM polarization and PMN-MDSC recruitment. Notably, targeting the CCL2/CCR2 or CXCL1/CXCR2 axis with the inhibitors RS504393 or Navarixin, respectively, effectively suppresses lung metastasis induced by RCC-derived C3 in a mouse model. Clinically, RCC patients with high expression of C3 demonstrate poor prognosis. Collectively, our findings reveal that tumor-derived EV C3 induces an immunosuppressive tumor microenvironment via TAMs, and thus promoting RCC metastasis.
Collapse
Affiliation(s)
- Yibi Zhang
- School of Biomedical Engineering (Suzhou), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei230026, China
- Key Laboratory of Bio-medical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou215163, China
- Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Southeast University, Nanjing210096, China
| | - Xiaodong Wang
- School of Biomedical Engineering (Suzhou), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei230026, China
- Key Laboratory of Bio-medical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou215163, China
- Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Southeast University, Nanjing210096, China
| | - Yinmin Gu
- Zhongda Hospital, Medical School, Advanced Institute for Life and Health, Southeast University, Nanjing210096, China
| | - Tongfeng Liu
- Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Southeast University, Nanjing210096, China
- Medical College, Guizhou University, Guiyang550025, China
| | - Xujie Zhao
- Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Southeast University, Nanjing210096, China
| | - Shuwen Cheng
- Medical School of Nanjing University, Nanjing210046, China
| | - Liqiang Duan
- Shanxi Academy of Advanced Research and Innovation, Shanxi Provincial Key Laboratory of Protein Structure Determination, Taiyuan030032, China
| | - Chang Huang
- Medical College, Guizhou University, Guiyang550025, China
| | - Songzhe Wu
- Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Southeast University, Nanjing210096, China
| | - Shan Gao
- Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Southeast University, Nanjing210096, China
| |
Collapse
|
29
|
Su B, Fan Z, Wu J, Zhan H. Genetic association of lipid-lowering drug target genes with pancreatic cancer: a Mendelian randomization study. Sci Rep 2025; 15:3282. [PMID: 39863728 PMCID: PMC11762976 DOI: 10.1038/s41598-025-87490-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Accepted: 01/20/2025] [Indexed: 01/27/2025] Open
Abstract
Previous studies have found that dyslipidemia is a risk factor for pancreatic cancer (PC), and that lipid-lowering drugs may reduce the risk of PC. However, it is not clear whether dyslipidemia causes PC. The Mendelian randomization (MR) study aimed to investigate the causal role of lipid traits in pancreatic cancer and to assess the potential impact of lipid-lowering drug targets on pancreatic cancer. Genetic variants associated with lipid traits and variants of genes encoding lipid-lowering drug targets were extracted from the Global Lipids Genetics Consortium genome-wide association study (GWAS). Summary statistics for PC were obtained from an independent GWAS datasets. Colocalization analyses were performed to validate the robustness of the results. No significant effect of lipid-lowering drug targets on PC risk was found. Genetic mimicry of lipoprotein lipase (LPL) was potentially associated with PC risks. Significant MR associations were observed in the discovery dataset (OR 1.64 [95% CI 1.24-2.16], p = 4.48*10-4) with PC in one dataset. However, the finding was not verified in the replication dataset. Our findings do not support dyslipidemia as a causal factor for PC. Among lipid-lowering drug targets, LPL is the potential drug target in PC.
Collapse
Affiliation(s)
- Bohan Su
- Division of Pancreatic Surgery, Department of General Surgery, Qilu Hospital, Shandong University, Jinan, 250012, China
| | - Zhiyao Fan
- Division of Pancreatic Surgery, Department of General Surgery, Qilu Hospital, Shandong University, Jinan, 250012, China
| | - Jiexi Wu
- Division of Pancreatic Surgery, Department of General Surgery, Qilu Hospital, Shandong University, Jinan, 250012, China
| | - Hanxiang Zhan
- Division of Pancreatic Surgery, Department of General Surgery, Qilu Hospital, Shandong University, Jinan, 250012, China.
| |
Collapse
|
30
|
Shen Y, Lin J, Jiang T, Shen X, Li Y, Fu Y, Xu P, Fang L, Chen Z, Huang H, Xia Y, Xu Z, Wang L. GC-derived exosomal circMAN1A2 promotes cancer progression and suppresses T-cell antitumour immunity by inhibiting FBXW11-mediated SFPQ degradation. J Exp Clin Cancer Res 2025; 44:24. [PMID: 39856764 PMCID: PMC11762487 DOI: 10.1186/s13046-025-03288-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Accepted: 01/13/2025] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND Exosomes, as extracellular membrane vesicles, play important roles in intercellular communication and can influence tumour progression. Circular RNAs (circRNAs) have been reported in various malignancies and are also important components of exosomes. However, the role of exosomal circRNAs in gastric cancer (GC) progression has not been completely clarified. METHODS The exosomal circRNAs enriched in GC were identified using exosomal circRNA sequencing. The biological function of circMAN1A2 in GC was investigated using a series of in vitro and in vivo experiments. PKH-67 staining was used to label the exosomes. The molecular mechanism of exosomal circMAN1A2 was investigated via mass spectrometry, immunoprecipitation, Western blot, and single-cell RNA-sequencing data analyses. RESULTS In our study, we determined that circMAN1A2 (hsa_circ_0000118) was enriched in GC-derived exosomes. Higher circMAN1A2 expression was related to poor survival in GC patients (HR = 2.917, p = 0.0120). Exosomal circMAN1A2 promoted GC progression in vitro and in vivo and suppressed the antitumour activity of T cells. Moreover, circMAN1A2 bound to SFPQ in GC cells and T cells, promoting the G1/S phase transition of the cell cycle in GC cells while inhibiting the activation of the T cell receptor signalling pathway in T cells to decrease antitumour activity. Mechanistically, circMAN1A2 competed with FBXW11 for binding to SFPQ, preventing FBXW11-mediated k48-linked ubiquitination and SFPQ protein degradation, thereby stabilizing SFPQ expression. CONCLUSIONS Our work confirms the critical role of exosomal circMAN1A2 in the progression and immunosuppression of GC. This novel axis of circMAN1A2-SFPQ provides new insights into exosomal circRNA-based GC diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- Yikai Shen
- Gastric Cancer Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Jie Lin
- Gastric Cancer Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Tianlu Jiang
- The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi Medical Center, Nanjing Medical University, Wuxi People's Hospital, Wuxi, Jiangsu Province, China
| | - Xusheng Shen
- Gastric Cancer Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Ying Li
- Gastric Cancer Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Yiwang Fu
- Gastric Cancer Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Penghui Xu
- Gastric Cancer Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Lang Fang
- Gastric Cancer Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Zetian Chen
- Gastric Cancer Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Hongxin Huang
- Gastric Cancer Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Yiwen Xia
- Gastric Cancer Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China.
| | - Zekuan Xu
- Gastric Cancer Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China.
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu Province, China.
| | - Linjun Wang
- Gastric Cancer Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China.
| |
Collapse
|
31
|
Simpson MA. Impacts of Hyaluronan on Extracellular Vesicle Production and Signaling. Cells 2025; 14:139. [PMID: 39851567 PMCID: PMC11763598 DOI: 10.3390/cells14020139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Revised: 01/14/2025] [Accepted: 01/15/2025] [Indexed: 01/26/2025] Open
Abstract
Hyaluronan (HA) is a critical component of cell and tissue matrices and an important signaling molecule. The enzymes that synthesize and process HA, as well as the HA receptors through which the signaling properties of HA are transmitted, have been identified in extracellular vesicles and implicated in context-specific processes associated with health and disease. The goal of this review is to present a comprehensive summary of the research on HA and its related receptors and enzymes in extracellular vesicle biogenesis and the cellular responses to vesicles bearing these extracellular matrix modulators. When present in extracellular vesicles, HA is assumed to be on the outside of the vesicle and is sometimes found associated with CD44 or the HAS enzyme itself. Hyaluronidases may be inside the vesicles or present on the vesicle surface via a transmembrane domain or GPI linkage. The implication of presenting these signals in extracellular vesicles is that there is a greater range of systemic distribution and more complex delivery media than previously thought for secreted HA or hyaluronidase alone. Understanding the context for these HA signals offers new diagnostic and therapeutic insight.
Collapse
Affiliation(s)
- Melanie A Simpson
- Department of Molecular and Structural Biochemistry, North Carolina State University, Raleigh, NC 27695-7622, USA
| |
Collapse
|
32
|
Sheth M, Sharma M, Kongsomros S, Lehn M, Takebe T, Takiar V, Wise-Draper T, Chutipongtanate S, Esfandiari L. Matrix stiffness modulated release of spheroid-derived extracellular vesicles and discovery of Piezo1 cargo. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.13.632826. [PMID: 39868119 PMCID: PMC11760731 DOI: 10.1101/2025.01.13.632826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
Augmented extracellular matrix (ECM) stiffness is a mechanical hallmark of cancer. Mechanotransduction studies have extensively probed the mechanisms by which ECM stiffness regulates intracellular communication. However, the influence of stiffness on intercellular communication aiding tumor progression in three-dimensional microenvironments remains unknown. Small extracellular vesicles (EVs) are communicators of altered biophysical cues to distant sites through EV-ECM interactions and EV-mediated recipient cell-ECM interactions. Here we demonstrate stiffness-mediated modulation of small EVs secretion and cargo from three-dimensional oral squamous cell carcinoma spheroids. Using a spheroid culture platform with varying matrix stiffness properties, we show that small EVs carry parental biomolecular cargo, including mechanosensitive Piezo1 ion channel and adhesion molecule CD44. We comprehensively validate the presence of both markers in our EV populations using proteomic and genetic analysis. Transcriptomic analysis of microRNA and long non-coding RNA cargo of small EVs released from soft and stiff ECM spheroids revealed enrichment of tumorigenic and metastatic profiles in EVs from stiff ECM cultures compared to that of soft ones. Gene set enrichment analysis of a comparative dataset obtained by overlaying spheroid mRNA and EV miRNA profiles identified key oncogenic pathways involved in cell-EV crosstalk in the spheroid model.
Collapse
|
33
|
Fan Y, Song S, Pizzi MP, Zou G, Vykoukal JV, Yoshimura K, Jin J, Calin GA, Waters RE, Gan Q, Wang L, Hanash S, Dhar SS, Ajani JA. Exosomal Galectin-3 promotes peritoneal metastases in gastric adenocarcinoma via microenvironment alterations. iScience 2025; 28:111564. [PMID: 39811647 PMCID: PMC11731617 DOI: 10.1016/j.isci.2024.111564] [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: 03/12/2024] [Revised: 06/22/2024] [Accepted: 12/06/2024] [Indexed: 01/16/2025] Open
Abstract
Peritoneal carcinomatosis (PC) in gastric adenocarcinoma (GAC) is the most common metastatic site and leads to a short median survival. Exosomes have been shown to remodel the microenvironment, facilitating tumor metastases. However, the functional component in GAC cell-derived exosomes that remodel the landscape in the peritoneal cavity remains unclear. To address this, we performed in-depth proteomic profiling of ascites-derived exosomes from patients with PC, and we found that Galectin-3 was highly enriched in exosomes derived from malignant ascites. exosomal Galectin-3 was the crucial regulator of PC. Blockage of exosomal Galectin-3 significantly inhibited tumor metastases and prolonged overall survival. Exosomal Galectin-3 activated cancer-associated fibroblasts through integrin α1β1/FAK/Akt/mTOR/CXCL12 signaling. Combined inhibition of the CXCL12-CXCR4 axis and exosomal Galectin-3 enhanced the efficacy of anti-PD-1 immunotherapy, leading to significantly diminished PC progression and durable antitumor responses. These findings provide a rationale for clinical strategy of targeting exosomal Galectin-3 to treat PC.
Collapse
Affiliation(s)
- Yibo Fan
- Department of Gastrointestinal Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Shumei Song
- Coriell Institute for Medical Research, Camden, NJ 08103, USA
| | - Melissa Pool Pizzi
- Department of Gastrointestinal Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Gengyi Zou
- Department of Gastrointestinal Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jody V Vykoukal
- Department of Clinical Cancer Prevention, Division of Cancer Prevention and Population Sciences, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Katsuhiro Yoshimura
- Department of Gastrointestinal Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Department of Tumor Pathology, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka 431-3192, Japan
| | - Jiankang Jin
- Department of Gastrointestinal Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - George A Calin
- Department of Translational Molecular Pathology, Division of Pathology and Laboratory Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Rebecca E Waters
- Department of Pathology, Division of Pathology and Laboratory Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Qiong Gan
- Department of Pathology, Division of Pathology and Laboratory Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Linghua Wang
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Samir Hanash
- Department of Clinical Cancer Prevention, Division of Cancer Prevention and Population Sciences, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Shilpa S Dhar
- Department of Gastrointestinal Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jaffer A Ajani
- Department of Gastrointestinal Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
34
|
Sojdeh S, Safarkhani M, Daneshgar H, Aldhaher A, Heidari G, Nazarzadeh Zare E, Iravani S, Zarrabi A, Rabiee N. Promising breakthroughs in amyotrophic lateral sclerosis treatment through nanotechnology's unexplored frontier. Eur J Med Chem 2025; 282:117080. [PMID: 39577228 DOI: 10.1016/j.ejmech.2024.117080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Revised: 11/15/2024] [Accepted: 11/16/2024] [Indexed: 11/24/2024]
Abstract
This review explores the transformative potential of nanotechnology in the treatment and diagnosis of amyotrophic lateral sclerosis (ALS), a progressive neurodegenerative disorder characterized by motor neuron degeneration, muscle weakness, and eventual paralysis. Nanotechnology offers innovative solutions across various domains, including targeted drug delivery, neuroprotection, gene therapy and editing, biomarker detection, advanced imaging techniques, and tissue engineering. By enhancing the precision and efficacy of therapeutic interventions, nanotechnology facilitates key advancements such as crossing the blood-brain barrier, targeting specific cell types, achieving sustained therapeutic release, and enabling combination therapies tailored to the complex pathophysiology of ALS. Despite its immense promise, the clinical translation of these approaches faces challenges, including potential cytotoxicity, biocompatibility, and regulatory compliance, which must be addressed through rigorous research and testing. This review emphasizes the application of nanotechnology in targeted drug delivery and gene therapy/editing for ALS, drawing on the author's prior work with various nanotechnological platforms to illustrate strategies for overcoming similar obstacles in drug and gene delivery. By bridging the gap between cutting-edge technology and clinical application, this article aims to highlight the vital role of nanotechnology in shaping the future of ALS treatment.
Collapse
Affiliation(s)
- Soheil Sojdeh
- Department of Chemistry, Sharif University of Technology, Tehran, Iran
| | - Moein Safarkhani
- Department of Biological Sciences and Bioengineering, Nano-Bio High-Tech Materials Research Center, Inha University, Incheon, 22212, Republic of Korea
| | - Hossein Daneshgar
- Department of Inorganic Chemistry, Faculty of Chemistry and Petroleum Sciences, Shahid Beheshti University, P. O. Box 19839-63113, Tehran, Iran
| | - Abdullah Aldhaher
- Department of Chemistry, Sharif University of Technology, Tehran, Iran
| | - Golnaz Heidari
- School of Natural Sciences, Massey University, Private Bag 11 222, Palmerston North, 4410, New Zealand
| | - Ehsan Nazarzadeh Zare
- School of Chemistry, Damghan University, Damghan, 36716-45667, Iran; Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, 140401, Punjab, India
| | | | - Ali Zarrabi
- Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Istinye University, 34396, Istanbul, Turkey; Graduate School of Biotechnology and Bioengineering, Yuan Ze University, Taoyuan, 320315, Taiwan
| | - Navid Rabiee
- Department of Biomaterials, Saveetha Dental College and Hospitals, SIMATS, Saveetha University, Chennai, 600077, India.
| |
Collapse
|
35
|
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
|
36
|
Zhang Y, Li B, Gu W, Fan L, Wang X, Xu M, Zhu M, Jin C. Hepatoma cell-derived exosomal SNORD52 mediates M2 macrophage polarization by activating the JAK2/STAT6 pathway. Discov Oncol 2025; 16:36. [PMID: 39804511 PMCID: PMC11730036 DOI: 10.1007/s12672-024-01700-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Accepted: 12/12/2024] [Indexed: 01/16/2025] Open
Abstract
BACKGROUND A recent study revealed the oncogenic role of box C/D small nucleolar RNA 52 (SNORD52) in hepatocellular carcinoma (HCC) by facilitating the aggressive phenotypes of hepatoma cells. However, the potential role of exosomal SNORD52 in macrophage polarization during HCC progression remains poorly understood. METHODS Exosomes were isolated from hepatoma cells. Western blotting and flow cytometry were performed to determine the levels of M2 macrophage polarization markers. SNORD52 expression was assessed using qRT-PCR. The levels of JAK2/STAT6 pathway-related proteins were analyzed using western blotting. RESULTS SNORD52 was enriched in exosomes derived from hepatoma cells and in plasma samples from patients with HCC. Hepatoma cell-derived exosomal SNORD52 was internalized by THP-1 macrophages. SNORD52 overexpression increased the levels of M2 macrophage polarization markers and JAK2/STAT6 pathway-related proteins Additionally, hepatoma cell-derived exosomal SNORD52 interacted with the JAK2/STAT6 pathway to mediate M2 macrophage polarization. CONCLUSIONS Our findings revealed that hepatoma cell-derived exosomal SNORD52 induces M2 macrophage polarization by activating the JAK2/STAT6 pathway.
Collapse
Affiliation(s)
- Yaqiong Zhang
- Department of Clinical Laboratory, Taizhou Central Hospital (Taizhou University Hospital), No.999 Donghai Road, Taizhou, 318000, Zhejiang, China
| | - Bo Li
- Department of Ultrasound, Taizhou Hospital, Zhejiang University, Taizhou Enze Medical Center (Group), Taizhou, 318000, Zhejiang, China
| | - Wanhong Gu
- Department of Clinical Laboratory, Taizhou Central Hospital (Taizhou University Hospital), No.999 Donghai Road, Taizhou, 318000, Zhejiang, China
| | - Linna Fan
- School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Xiaofan Wang
- Department of Clinical Laboratory, Taizhou Central Hospital (Taizhou University Hospital), No.999 Donghai Road, Taizhou, 318000, Zhejiang, China
| | - Meifen Xu
- Department of Clinical Laboratory, Taizhou Central Hospital (Taizhou University Hospital), No.999 Donghai Road, Taizhou, 318000, Zhejiang, China
| | - Minqi Zhu
- Department of Clinical Laboratory, Taizhou Central Hospital (Taizhou University Hospital), No.999 Donghai Road, Taizhou, 318000, Zhejiang, China.
| | - Chong Jin
- Department of Hepatobiliary Pancreatic Splenic Surgery, Taizhou Central Hospital (Taizhou University Hospital), No.999 Donghai Road, Taizhou, 318000, Zhejiang, China.
| |
Collapse
|
37
|
Xie J, Lin X, Deng X, Tang H, Zou Y, Chen W, Xie X. Cancer-associated fibroblast-derived extracellular vesicles: regulators and therapeutic targets in the tumor microenvironment. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2025; 8:2. [PMID: 39935427 PMCID: PMC11810458 DOI: 10.20517/cdr.2024.152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 12/19/2024] [Accepted: 12/31/2024] [Indexed: 02/13/2025]
Abstract
Cancer-associated fibroblasts (CAFs) constitute a critical component of the tumor microenvironment (TME). CAFs can be reprogrammed by cancer cells, leading to the production of extracellular vesicles (EVs). These EVs serve as carriers for bioactive substances, including proteins, nucleic acids, and metabolic products, thereby facilitating tumor progression. CAF-derived EVs exert substantial influence on tumor cell proliferation, invasion, and metastasis, the immunological environment, and the processes of lymphangiogenesis and angiogenesis. Despite their potential as non-invasive biomarkers and therapeutic delivery vehicles, the clinical application of CAF-derived EVs is currently limited by challenges in purification and precise targeting. This review delineates the diverse roles of CAF-derived EVs in tumor growth, metastasis, and immune evasion within the TME.
Collapse
Affiliation(s)
- Jindong Xie
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, Guangdong, China
- Authors contributed equally
| | - Xinmei Lin
- School of Medicine, Sun Yat-sen University, Guangzhou 510080, Guangdong, China
- Authors contributed equally
| | - Xinpei Deng
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, Guangdong, China
- Authors contributed equally
| | - Hailin Tang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, Guangdong, China
| | - Yutian Zou
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, Guangdong, China
| | - Wenkuan Chen
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, Guangdong, China
| | - Xiaoming Xie
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, Guangdong, China
| |
Collapse
|
38
|
Choi BM, Park TJ, Lee HH, Hong H, Chi WJ, Kim SY. Inhibition of Melanin Synthesis and Inflammation by Exosomes Derived from Leuconostoc mesenteroides DB-14 Isolated from Camellia japonica Flower. J Microbiol Biotechnol 2025; 35:e2411080. [PMID: 39809510 PMCID: PMC11813340 DOI: 10.4014/jmb.2411.11080] [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: 11/30/2024] [Revised: 12/20/2024] [Accepted: 12/27/2024] [Indexed: 01/16/2025]
Abstract
Leuconostoc mesenteroides is a lactic acid bacteria found in fermented products. In our previous study, L. mesenteroides was isolated from Camellia japonica flowers, and its acid tolerance and antibacterial properties were thoroughly investigated. This study focuses on the inhibition of melanin synthesis and inflammation of exosomes derived from L. mesenteroides. Moreover, L. mesenteroides exosomes (DB-14 exosome) exhibited significant inhibitory effects on inflammation and melanogenesis. At concentrations of 4.44 × 108, 8.88 × 108, and 1.78 × 109 particles/ml, the exosomes reduced nitric oxide and prostaglandin E2 activity while maintaining the growth of RAW 264.7 macrophages. In addition, proinflammatory cytokines, such as interleukin (IL)-1β, IL-6, and tumor necrosis factor alpha, were rarely expressed, and western blot revealed that L. mesenteroides DB-14 derived exosomes inhibited inducible nitric oxide synthase and cyclooxygenase-2 expression. Moreover, the exosomes had no toxic effects on B16F10 melanoma cells at concentrations of 1.78 × 109, 3.55 × 109, and 7.10 × 109 particles/ml, and they suppressed melanogenesis by reducing tyrosinase activity. Furthermore, western blot analysis demonstrated that microphthalmia-associated transcription factor (MITF), tyrosinase, tyrosinase related protein (TRP)-1, and TRP-2 were evidently reduced, ultimately repressing melanin production. Moreover, MITF expression was inhibited by reduced mitogen-activated protein kinase and protein kinase B phosphorylation levels. Overall, this study proves the efficacy of the novel DB-14 exosome as a strong lightening and anti-inflammatory agent.
Collapse
Affiliation(s)
- Byeong Min Choi
- Department of Pharmaceutical Engineering & Biotechnology, Sunmoon University, Chungnam 31460, Republic of Korea
| | - Tae-Jin Park
- Department of Pharmaceutical Engineering & Biotechnology, Sunmoon University, Chungnam 31460, Republic of Korea
| | | | - Hyehyun Hong
- Department of Pharmaceutical Engineering & Biotechnology, Sunmoon University, Chungnam 31460, Republic of Korea
| | - Won-Jae Chi
- Biodiversity Research Department, Species Diversity Research Division, Incheon 22689, Republic of Korea
| | - Seung-Young Kim
- Department of Pharmaceutical Engineering & Biotechnology, Sunmoon University, Chungnam 31460, Republic of Korea
| |
Collapse
|
39
|
Luan X, Wang X, Bian G, Li X, Gao Z, Liu Z, Zhang Z, Han T, Zhao J, Zhao H, Luan X, Zhu W, Dong L, Guo F. Exosome applications for the diagnosis and treatment of pancreatic ductal adenocarcinoma: An update (Review). Oncol Rep 2025; 53:13. [PMID: 39575479 PMCID: PMC11605277 DOI: 10.3892/or.2024.8846] [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: 08/08/2024] [Accepted: 10/30/2024] [Indexed: 11/28/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a malignant neoplasm that typically manifests with subtle clinical manifestations in its early stages and frequently eludes diagnosis until the advanced phases of the disease. The limited therapeutic options available for PDAC significantly contribute to its high mortality rate, highlighting the urgent need for novel biomarkers capable of effectively identifying early clinical manifestations and facilitating precise diagnosis. The pivotal role of cellular exosomes in both the pathogenesis and therapeutic interventions for PDAC has been underscored. Furthermore, researchers have acknowledged the potential of exosomes as targeted drug carriers against regulatory cells in treating PDAC. The present article aims to provide a comprehensive review encompassing recent advancements in utilizing exosomes for elucidating mechanisms underlying disease development, patterns of metastasis, diagnostic techniques and treatment strategies associated with PDAC.
Collapse
Affiliation(s)
- Xinchi Luan
- Pathophysiology Department, School of Basic Medicine, Qingdao University, Qingdao, Shandong 266071, P.R. China
| | - Xuezhe Wang
- Pathophysiology Department, School of Basic Medicine, Qingdao University, Qingdao, Shandong 266071, P.R. China
| | - Gang Bian
- Department of Gastroenterology, Affiliated Qingdao Third People's Hospital, Qingdao University, Qingdao, Shandong 266041, P.R. China
| | - Xiaoxuan Li
- Department of Oncology, Key Laboratory of Cancer Molecular and Translational Research, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266031, P.R. China
| | - Ziru Gao
- Pathophysiology Department, School of Basic Medicine, Qingdao University, Qingdao, Shandong 266071, P.R. China
| | - Zijiao Liu
- School of Clinical and Basic Medicine and Institute of Basic Medicine, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, P.R. China
| | - Zhishang Zhang
- Pathophysiology Department, School of Basic Medicine, Qingdao University, Qingdao, Shandong 266071, P.R. China
| | - Tianyue Han
- Pathophysiology Department, School of Basic Medicine, Qingdao University, Qingdao, Shandong 266071, P.R. China
| | - Jinpeng Zhao
- Pathophysiology Department, School of Basic Medicine, Qingdao University, Qingdao, Shandong 266071, P.R. China
| | - Hongjiao Zhao
- Pathophysiology Department, School of Basic Medicine, Qingdao University, Qingdao, Shandong 266071, P.R. China
| | - Xinyue Luan
- Pathophysiology Department, School of Basic Medicine, Qingdao University, Qingdao, Shandong 266071, P.R. China
| | - Wuhui Zhu
- Department of Hepatobiliary surgery, Affiliated Qingdao Third People's Hospital, Qingdao University, Qingdao, Shandong 266041, P.R. China
| | - Lili Dong
- Department of Gastroenterology, Affiliated Qingdao Third People's Hospital, Qingdao University, Qingdao, Shandong 266041, P.R. China
| | - Feifei Guo
- Pathophysiology Department, School of Basic Medicine, Qingdao University, Qingdao, Shandong 266071, P.R. China
| |
Collapse
|
40
|
Zhang H, Wu J, Gan J, Wang W, Liu Y, Song T, Yang Y, Ji G, Li W. Proteomic Analysis of Plasma Exosomes Enables the Identification of Lung Cancer in Patients With Chronic Obstructive Pulmonary Disease. Thorac Cancer 2025; 16:e15517. [PMID: 39778061 PMCID: PMC11717053 DOI: 10.1111/1759-7714.15517] [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: 10/18/2024] [Revised: 11/30/2024] [Accepted: 12/09/2024] [Indexed: 01/11/2025] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is confirmed as an independent risk factor for the development of lung cancer. Although low-dose CT screening significantly reduces the mortality rate of lung cancer, the misdiagnosis and missed diagnosis rates remain high in the COPD population. Additionally, several COPD patients are unable to undergo invasive histological examinations. Therefore, there is an urgent need for minimally invasive biomarkers to screen or diagnose lung cancer in COPD patients. In this study, peripheral blood samples were collected from COPD patients with and without lung cancer. Plasma exosomes (EVs) were extracted for proteomic analysis. Sixteen differentially expressed proteins (DEPs) were preliminarily selected via label-free quantification (LFQ) proteomic technology and comprehensive bioinformatics analysis. Parallel reaction monitoring (PRM) targeted validation identified five candidate proteins associated with COPD with lung cancer. Compared to the COPD group, KRT1, KRT9, and KRT10 were significantly upregulated in the COPD with lung cancer group, while GPLD1 and TF were downregulated. The biomarkers identified in our study provide a foundation for non-invasive screening and diagnosis of lung cancer in COPD patients and exploration of the mechanisms shared between COPD and lung cancer.
Collapse
Affiliation(s)
- Huohuo Zhang
- Department of Pulmonary and Critical Care Medicine, West China HospitalSichuan UniversityChengduSichuanChina
- Institute of Respiratory Health and Multimorbidity, West China HospitalSichuan UniversityChengduSichuanChina
| | - Jiaxuan Wu
- Department of Pulmonary and Critical Care Medicine, West China HospitalSichuan UniversityChengduSichuanChina
- Institute of Respiratory Health and Multimorbidity, West China HospitalSichuan UniversityChengduSichuanChina
| | - Jiadi Gan
- Department of Pulmonary and Critical Care Medicine, West China HospitalSichuan UniversityChengduSichuanChina
- Institute of Respiratory Health and Multimorbidity, West China HospitalSichuan UniversityChengduSichuanChina
| | - Wei Wang
- Health Management Center, General Practice Medical Center, West China HospitalSichuan UniversityChengduSichuanChina
| | - Yi Liu
- Department of Pulmonary and Critical Care Medicine, West China HospitalSichuan UniversityChengduSichuanChina
- Institute of Respiratory Health and Multimorbidity, West China HospitalSichuan UniversityChengduSichuanChina
| | - Tingting Song
- Institute of Respiratory Health and Multimorbidity, West China HospitalSichuan UniversityChengduSichuanChina
- Precision Medicine Center, Precision Medicine Key Laboratory of Sichuan Province, West China HospitalSichuan UniversityChengduSichuanChina
- Institute of Respiratory Health, Frontiers Science Center for Disease‐Related Molecular Network, West China HospitalSichuan UniversityChengduSichuanChina
| | - Yongfeng Yang
- Institute of Respiratory Health and Multimorbidity, West China HospitalSichuan UniversityChengduSichuanChina
- Precision Medicine Center, Precision Medicine Key Laboratory of Sichuan Province, West China HospitalSichuan UniversityChengduSichuanChina
- Institute of Respiratory Health, Frontiers Science Center for Disease‐Related Molecular Network, West China HospitalSichuan UniversityChengduSichuanChina
| | - Guiyi Ji
- Health Management Center, General Practice Medical Center, West China HospitalSichuan UniversityChengduSichuanChina
| | - Weimin Li
- Department of Pulmonary and Critical Care Medicine, West China HospitalSichuan UniversityChengduSichuanChina
- Institute of Respiratory Health and Multimorbidity, West China HospitalSichuan UniversityChengduSichuanChina
- Precision Medicine Center, Precision Medicine Key Laboratory of Sichuan Province, West China HospitalSichuan UniversityChengduSichuanChina
- Institute of Respiratory Health, Frontiers Science Center for Disease‐Related Molecular Network, West China HospitalSichuan UniversityChengduSichuanChina
- The Research Units of West China, Chinese Academy of Medical SciencesWest China HospitalChengduSichuanChina
- State Key Laboratory of Respiratory Health and MultimorbidityWest China HospitalChengduSichuanChina
| |
Collapse
|
41
|
Cui W, Qu L, Xu Y, Wang Z, Gu Y, Tian S, Qi F, Pan H. MXene@Ni 3(HITP) 2@AuNPs combined with NiCo@Fc-MWCNTs-LDH for electrochemical detection of extracellular vesicles. Talanta 2025; 282:126987. [PMID: 39383717 DOI: 10.1016/j.talanta.2024.126987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 09/27/2024] [Accepted: 10/03/2024] [Indexed: 10/11/2024]
Abstract
This study proposed an electrochemical sensor combining Mxene@Ni3(HITP)2@AuNPs with NiCo@Fc-MWCNTs-LDH for detecting extracellular vesicles (EVs) derived from MCF-7 cells. Mxene exhibits high conductivity and large surface area. Ni3(HITP)2 is a novel conductive metal-organic framework (MOF) with outstanding conductivity, capable of loading more gold nanoparticles (AuNPs) when combined with polyetherimide (PEI). Tetrahedra DNA (TDN) is anchored on the substrate through gold nanoparticles (AuNPs) for the specific capture of EVs, with CD63 aptamers carried at their vertices. In the signal layer, the NiCo@Fc-MWCNTs-LDH loaded with CD63 aptamers was prepared as the electrochemical sensor signal label for EVs detection. This electrochemical sensor exhibits high sensitivity, evidenced by a low limit of detection (LOD) of 13.79 particles/mL and a linear range from 1.6 × 102 to 1.6 × 106 particles/mL, underscoring its potential for early cancer diagnosis.
Collapse
Affiliation(s)
- Weikang Cui
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, 200093, China; Shanghai University of Medicine and Health Sciences, Shanghai, 201318, China
| | - Lingli Qu
- Shanghai Urban Construction Vocational College, Shanghai, 201999, China.
| | - Yiwen Xu
- Shanghai University of Medicine and Health Sciences, Shanghai, 201318, China
| | - Zekai Wang
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, 200093, China; Shanghai University of Medicine and Health Sciences, Shanghai, 201318, China
| | - Yangyang Gu
- Shanghai University of Medicine and Health Sciences, Shanghai, 201318, China
| | - Siyue Tian
- Shanghai University of Medicine and Health Sciences, Shanghai, 201318, China
| | - Feifan Qi
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, 200093, China; Shanghai University of Medicine and Health Sciences, Shanghai, 201318, China
| | - Hongzhi Pan
- Shanghai University of Medicine and Health Sciences, Shanghai, 201318, China.
| |
Collapse
|
42
|
Kasherwal V, Kale V, Vaidya A. Extracellular vesicles secreted by leukemic cells as mediators of dysregulated hematopoiesis: acute myeloid leukemia as a case in point. Expert Rev Hematol 2025; 18:225-237. [PMID: 40008450 DOI: 10.1080/17474086.2025.2471860] [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: 10/26/2024] [Revised: 01/23/2025] [Accepted: 02/19/2025] [Indexed: 02/27/2025]
Abstract
INTRODUCTION Acute myeloid leukemia (AML) cells exhibit a profound capacity for resistance to conventional chemotherapeutic agents, posing a substantial challenge to existing therapeutic paradigms. Interestingly, this happens in the face of a luxuriant proliferation of leukemic blasts in the peripheral blood. This paradox of concurrent proliferative activity and cellular quiescence underscores a complex biological phenomenon that is intricately mediated by AML-derived Extracellular vesicles (EVs). AREAS COVERED An extensive literature review search was done on PubMed/Scopus/Web of Sciences databases to identify studies published between 2013 and 2024 elucidating and demonstrating the effect of AML-derived EVs, Microvesicles (MVs) and Exosomes (Exos) in regulating the normal and dysregulated bone marrow (BM) niche. EXPERT OPINION The review delves into understanding the molecular mechanisms underlying the dual behavior of AML cells - proliferation and quiescence, with a special focus on the role of the EVs and their subtypes viz. Exos and MVs in establishing a discrete BM microenvironment that is subversive to chemotherapy. It offers a novel perspective on the intricate interplay between the leukemic cells and their microenvironment, with implications for therapeutic interventions targeting AML persistence and drug resistance.
Collapse
Affiliation(s)
- Vishakha Kasherwal
- Symbiosis School of Biological Sciences, Symbiosis International (Deemed University), Pune, India
| | - Vaijayanti Kale
- Symbiosis Centre for Stem Cell Research, Symbiosis International (Deemed University), Pune, India
| | - Anuradha Vaidya
- Symbiosis School of Biological Sciences, Symbiosis International (Deemed University), Pune, India
- Symbiosis Centre for Stem Cell Research, Symbiosis International (Deemed University), Pune, India
| |
Collapse
|
43
|
Fan X, Lin J, Liu H, Deng Q, Zheng Y, Wang X, Yang L. The role of macrophage-derived exosomes in noncancer liver diseases: From intercellular crosstalk to clinical potential. Int Immunopharmacol 2024; 143:113437. [PMID: 39454408 DOI: 10.1016/j.intimp.2024.113437] [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: 10/07/2024] [Accepted: 10/16/2024] [Indexed: 10/28/2024]
Abstract
Chronic liver disease has a substantial global prevalence and mortality rate. Macrophages, pivotal cells in innate immunity, exhibit remarkable heterogeneity and plasticity and play a considerable role in maintaining organ homeostasis, modulating inflammatory responses, and influencing disease progression in the liver. Exosomes, which can serve as conduits for intercellular communication, biomarkers, and therapeutic targets for a spectrum of diseases, have recently garnered increasing attention recently. Given that the liver is the organ with the highest macrophage content, a thorough understanding of the influence of macrophage-derived exosomes (MDEs) on noncancer liver disease pathogenesis and their potential therapeutic applications is paramount. Interactions among MDEs, hepatocytes, hepatic stellate cells (HSCs), and other nonparenchymal cells constitute a complex network regulates liver immune homeostasis. In this review, we summarize the latest progress in the current understanding of MDE heterogeneity and cellular crosstalk in noncancer liver diseases, as well as their potential clinical applications. Additionally, challenges and future directions are underscored.
Collapse
Affiliation(s)
- Xiaoli Fan
- Department of Gastroenterology and Hepatology and Laboratory of Gastrointestinal Cancer and Liver Disease, West China Hospital, Sichuan University, Chengdu, China
| | - Jin Lin
- Department of Gastroenterology and Hepatology and Laboratory of Gastrointestinal Cancer and Liver Disease, West China Hospital, Sichuan University, Chengdu, China
| | - Honglan Liu
- Dazhou Central Hospital, Dazhou 635000, Sichuan Province, China
| | - Qiaoyu Deng
- Department of Gastroenterology and Hepatology and Laboratory of Gastrointestinal Cancer and Liver Disease, West China Hospital, Sichuan University, Chengdu, China
| | - Yanyi Zheng
- Department of Gastroenterology and Hepatology and Laboratory of Gastrointestinal Cancer and Liver Disease, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaoze Wang
- Department of Gastroenterology and Hepatology and Laboratory of Gastrointestinal Cancer and Liver Disease, West China Hospital, Sichuan University, Chengdu, China.
| | - Li Yang
- Department of Gastroenterology and Hepatology and Laboratory of Gastrointestinal Cancer and Liver Disease, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
44
|
Abida, Alhuthali HM, Alshehri JM, Alkathiri A, Almaghrabi ROM, Alsaeed SS, Albebi SAH, Almethn RM, Alfuraydi BA, Alharbi SB, Kamal M, Imran M. Exosomes in infectious diseases: insights into leishmaniasis pathogenesis, immune modulation, and therapeutic potential. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024:10.1007/s00210-024-03702-7. [PMID: 39702600 DOI: 10.1007/s00210-024-03702-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Accepted: 12/02/2024] [Indexed: 12/21/2024]
Abstract
Leishmaniasis continues to be a critical international health issue due to the scarcity of efficient treatment and the development of drug tolerance. New developments in the research of extracellular vesicles (EVs), especially exosomes, have revealed novel disease management approaches. Exosomes are small vesicles that transport lipids, nucleic acids, and proteins in cell signalling. Its biogenesis depends on several cellular processes, and their functions in immune response, encompassing innate and adaptive immunity, underline their function in the pathogen-host interface. Exosomes play a significant role in the pathogenesis of some parasitic infections, especially Leishmaniasis, by helping parasites escape host immunity and promote disease progression. This article explains that in the framework of parasitic diseases, exosomes can act as master regulators that define the pathogenesis of the disease, as illustrated by the engagement of exosomes in the Leishmaniasis parasite and immune escape processes. Based on many published articles on Leishmaniasis, this review aims to summarize the biogenesis of exosomes, the properties of the cargo in exosomes, and the modulation of immune responses. We delve deeper into the prospect of using exosomes for the therapy of Leishmaniasis based on the possibility of using these extracellular vesicles for drug delivery and as diagnostic and prognostic biomarkers. Lastly, we focus on the recent research perspectives and future developments, underlining the necessity to continue the investigation of exosome-mediated approaches in Leishmaniasis treatment. Thus, this review intends to draw attention to exosomes as a bright new perspective in the battle against this disabling affliction.
Collapse
Affiliation(s)
- Abida
- Department of Pharmaceutical Chemistry, College of Pharmacy, Northern Border University, 91911, Rafha, Saudi Arabia
| | - Hayaa M Alhuthali
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, P.O. Box 11099, 21944, Taif, Saudi Arabia
| | - Jawaher Mohammad Alshehri
- Optometry Department, Faculty of Applied Medical Sciences, Albaha University, 65431, Albaha, Saudi Arabia
| | - Afnan Alkathiri
- Laboratory Medicine Department, Faculty of Applied Medical Sciences, Albaha University, 65431, Albaha, Saudi Arabia
| | - Ruba Omar M Almaghrabi
- Laboratory Medicine Department, Faculty of Applied Medical Sciences, Albaha University, 65431, Albaha, Saudi Arabia
| | | | | | | | | | | | - Mehnaz Kamal
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam Bin Abdulaziz University, 11942, Al-Kharj, Saudi Arabia
| | - Mohd Imran
- Department of Pharmaceutical Chemistry, College of Pharmacy, Northern Border University, 91911, Rafha, Saudi Arabia.
- Center for Health Research, Northern Border University, Arar, Saudi Arabia.
| |
Collapse
|
45
|
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
|
46
|
Wen F, Han Y, Zhang H, Zhao Z, Wang W, Chen F, Qin W, Ju J, An L, Meng Y, Yang J, Tang Y, Zhao Y, Zhang H, Li F, Bai W, Xu Y, Zhou Z, Jiao S. Epstein-Barr virus infection upregulates extracellular OLFM4 to activate YAP signaling during gastric cancer progression. Nat Commun 2024; 15:10543. [PMID: 39627192 PMCID: PMC11615309 DOI: 10.1038/s41467-024-54850-6] [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: 09/05/2023] [Accepted: 11/24/2024] [Indexed: 12/06/2024] Open
Abstract
Extracellular vesicles (EVs) are known to mediate cell communications and shape tumor microenvironment. Compared to the well-studied small EVs, the function of large microvesicles (MVs) during tumorigenesis is poorly understood. Here we show the proteome of MVs in Epstein-Barr virus (EBV)-associated gastric cancer (EBVaGC), and identify olfactomedin 4 (OLFM4) is induced by EBV infection and secreted via MVs to promote tumor progression through Hippo signaling. Specifically, OLFM4 is a target gene of the cGAS-STING pathway, and EBV infection activates cGAS-STING pathway and increases OLFM4 expression. Moreover, MV-carried OLFM4 binds with the extracellular cadherin domain of FAT1, thereby impairing its intracellular interaction with MST1 and leading to YAP activation in recipient cells. Together, our study not only reveals a regulatory mechanism though which viral infection is coupled via MVs with intercellular control of the Hippo signaling, but also highlights the OLFM4-Hippo axis as a therapeutic target for EBV-associated cancers.
Collapse
Affiliation(s)
- Fuping Wen
- Department of Stomatology, Shanghai Tenth People's Hospital, Tongji University Cancer Center, Tongji University School of Medicine, Shanghai, 200072, China
| | - Yi Han
- Department of Stomatology, Shanghai Tenth People's Hospital, Tongji University Cancer Center, Tongji University School of Medicine, Shanghai, 200072, China
| | - Hui Zhang
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai, 200438, China
| | - Zhangting Zhao
- Department of Stomatology, Shanghai Tenth People's Hospital, Tongji University Cancer Center, Tongji University School of Medicine, Shanghai, 200072, China
| | - Wenjia Wang
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai, 200438, China
| | - Fan Chen
- CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Weimin Qin
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai, 200438, China
| | - Junyi Ju
- Department of Stomatology, Shanghai Tenth People's Hospital, Tongji University Cancer Center, Tongji University School of Medicine, Shanghai, 200072, China
| | - Liwei An
- Department of Stomatology, Shanghai Tenth People's Hospital, Tongji University Cancer Center, Tongji University School of Medicine, Shanghai, 200072, China
| | - Yan Meng
- Department of Stomatology, Shanghai Tenth People's Hospital, Tongji University Cancer Center, Tongji University School of Medicine, Shanghai, 200072, China
| | - Jie Yang
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai, 200438, China
| | - Yang Tang
- Department of Stomatology, Shanghai Tenth People's Hospital, Tongji University Cancer Center, Tongji University School of Medicine, Shanghai, 200072, China
| | - Yun Zhao
- CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Huanhu Zhang
- Department of Digestive Sciences, Shanxi Cancer Hospital, Taiyuan, 030001, China
| | - Feng Li
- Department of Digestive Sciences, Shanxi Cancer Hospital, Taiyuan, 030001, China
| | - Wenqi Bai
- Department of Digestive Sciences, Shanxi Cancer Hospital, Taiyuan, 030001, China.
| | - Yuanzhi Xu
- Department of Stomatology, Shanghai Tenth People's Hospital, Tongji University Cancer Center, Tongji University School of Medicine, Shanghai, 200072, China.
| | - Zhaocai Zhou
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai, 200438, China.
- Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, 211166, China.
| | - Shi Jiao
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai, 200438, China.
| |
Collapse
|
47
|
Dilsiz N. A comprehensive review on recent advances in exosome isolation and characterization: Toward clinical applications. Transl Oncol 2024; 50:102121. [PMID: 39278189 PMCID: PMC11418158 DOI: 10.1016/j.tranon.2024.102121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Revised: 09/03/2024] [Accepted: 09/11/2024] [Indexed: 09/18/2024] Open
Abstract
Exosomes are small, round vesicles in the 30 and 120 nm diameter range released by all living cell types. Exosomes play many essential functions in intercellular communication and tissue crosstalk in the human body. They can potentially be used as strong biomarkers and therapeutic agents for early diagnosis, therapy response, and prognosis of different diseases. The main requirements for exosomal large-scale clinical practice application are rapid, easy, high-yield, high purity, characterization, safety, low cost, and therapeutic efficacy. Depending on the sample types, environmental insults, and exosome quantity, exosomes can be isolated from various sources, including body fluids, solid tissues, and cell culture medium using different procedures. This study comprehensively analyzed the current research progress in exosome isolation and characterization strategies along with their advantages and disadvantages. The provided information will make it easier to select exosome separation methods based on the types of biological samples available, and it will facilitate the use of exosomes in translational and clinical research, particularly in cancer. Lay abstract Exosomes have recently received much attention due to their potential to function as biomarkers and novel therapeutic agents for early diagnosis, therapeutic response, and prognosis in various diseases. This review summarizes many approaches for isolating and characterizing exosomes, focusing on developing technologies, and provides an in-depth comparison and analysis of each method, including its principles, advantages, and limitations.
Collapse
Affiliation(s)
- Nihat Dilsiz
- Experimental Medicine Application and Research Center (EMARC) Validebag Research Park, University of Health Sciences, Istanbul, Turkey.
| |
Collapse
|
48
|
Rak J. Anti-metastatic extracellular vesicles carrying DNA. NATURE CANCER 2024; 5:1793-1795. [PMID: 39627553 DOI: 10.1038/s43018-024-00829-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Affiliation(s)
- Janusz Rak
- McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
49
|
Jiang P, Ma X, Wang X, Huang J, Wang Y, Ai J, Xiao H, Dai M, Lin Y, Shao B, Tang X, Tong W, Ye Z, Chai R, Zhang S. Isolation and Comprehensive Analysis of Cochlear Tissue-Derived Small Extracellular Vesicles. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2408964. [PMID: 39497619 DOI: 10.1002/advs.202408964] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 10/19/2024] [Indexed: 12/28/2024]
Abstract
Small extracellular vesicles (sEVs) act as a critical mediator in intercellular communication. Compared to sEVs derived from in vitro sources, tissue-derived sEVs can reflect the in vivo signals released from specific tissues more accurately. Currently, studies on the role of sEVs in the cochlea have relied on studying sEVs from in vitro sources. This study evaluates three cochlear tissue digestion and cochlear tissue-derived sEV (CDsEV) isolation methods, and first proposes that the optimal approach for isolating CDsEVs using collagenase D and DNase І combined with sucrose density gradient centrifugation. Furthermore, it comprehensively investigates CDsEV contents and cell origins. Small RNA sequencing and proteomics are performed to analyze the miRNAs and proteins of CDsEVs. The miRNAs and proteins of CDsEVs are crucial for maintaining normal auditory function. Among them, FGFR1 in CDsEVs may mediate the survival of cochlear hair cells via sEVs. Finally, the joint analysis of single CDsEV sequencing and single-cell RNA sequencing data is utilized to trace cellular origins of CDsEVs. The results show that different types of cochlear cells secrete different amounts of CDsEVs, with Kölliker's organ cells and supporting cells secrete the most. The findings are expected to enhance the understanding of CDsEVs in the cochlea.
Collapse
Affiliation(s)
- Pei Jiang
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
- Southeast University Shenzhen Research Institute, Shenzhen, 1518063, China
| | - Xiangyu Ma
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
| | - Xinlin Wang
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
| | - Jingyuan Huang
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
| | - Yintao Wang
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
| | - Jingru Ai
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
- Southeast University Shenzhen Research Institute, Shenzhen, 1518063, China
| | - Hairong Xiao
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
- Southeast University Shenzhen Research Institute, Shenzhen, 1518063, China
| | - Mingchen Dai
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
| | - Yanqin Lin
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
- Southeast University Shenzhen Research Institute, Shenzhen, 1518063, China
| | - Buwei Shao
- School of Medicine, Faculty of Medical & Health Sciences, Tel Aviv University, Tel Aviv, 6997801, Israel
| | - Xujun Tang
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
| | - Wei Tong
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
| | - Zixuan Ye
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
- Southeast University Shenzhen Research Institute, Shenzhen, 1518063, China
| | - Renjie Chai
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
- Department of Otolaryngology Head and Neck Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China
- Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Science, Beijing, 100101, China
| | - Shasha Zhang
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
| |
Collapse
|
50
|
Lee SY, Klingeborn M, Bulte JWM, Chiu DT, Chopp M, Cutler CW, Das S, Egwuagu CE, Fowler CD, Hamm‐Alvarez SF, Lee H, Liu Y, Mead B, Moore TL, Ravindran S, Shetty AK, Skog J, Witwer KW, Djalilian AR, Weaver AM. A perspective from the National Eye Institute Extracellular Vesicle Workshop: Gaps, needs, and opportunities for studies of extracellular vesicles in vision research. J Extracell Vesicles 2024; 13:e70023. [PMID: 39665315 PMCID: PMC11635481 DOI: 10.1002/jev2.70023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 11/20/2024] [Accepted: 11/24/2024] [Indexed: 12/13/2024] Open
Abstract
With an evolving understanding and new discoveries in extracellular vesicle (EV) biology and their implications in health and disease, the significant diagnostic and therapeutic potential of EVs for vision research has gained recognition. In 2021, the National Eye Institute (NEI) unveiled its Strategic Plan titled 'Vision for the Future (2021-2025),' which listed EV research as a priority within the domain of Regenerative Medicine, a pivotal area outlined in the Plan. In alignment with this prioritization, NEI organized a workshop inviting twenty experts from within and beyond the visual system. The workshop aimed to review current knowledge in EV research and explore gaps, needs and opportunities for EV research in the eye, including EV biology and applications of EVs in diagnosis, therapy and prognosis within the visual system. This perspective encapsulates the workshop's deliberations, highlighting the current landscape and potential implications of EV research in advancing eye health and addressing visual diseases.
Collapse
Affiliation(s)
- Sun Young Lee
- Department of Ophthalmology, USC Roski Eye Institute, Keck School of MedicineUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | | | - Jeff W. M. Bulte
- Department of Radiology and Radiological Sciences, School of MedicineJohns Hopkins UniversityBaltimoreMarylandUSA
| | - Daniel T. Chiu
- Department of Chemistry and BioengineeringUniversity of WashingtonSeattleWashingtonUSA
| | - Michael Chopp
- Department of NeurologyHenry Ford HealthDetroitMichiganUSA
| | | | - Saumya Das
- Cardiovascular Research CenterMassachusetts General Hospital and Harvard Medical SchoolBostonMassachusettsUSA
| | - Charles E. Egwuagu
- Molecular Immunology Section, Laboratory of Immunology, National Eye InstituteNational Institutes of HealthBethesdaMarylandUSA
| | - Christie D. Fowler
- Department of Neurobiology and BehaviorUniversity of California IrvineIrvineCaliforniaUSA
| | - Sarah F. Hamm‐Alvarez
- Department of Ophthalmology, USC Roski Eye Institute, Keck School of MedicineUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Hakho Lee
- Center for System BiologyMassachusetts General Hospital and Harvard Medical SchoolBostonMassachusettsUSA
| | - Yutao Liu
- Department of Cellular Biology and Anatomy, Medical College of GeorgiaAugusta UniversityAugustaGeorgiaUSA
| | - Ben Mead
- School of Optometry and Vision SciencesCardiff UniversityCardiffUK
| | - Tara L. Moore
- Department of Anatomy and NeurobiologyBoston University Chobanian & Avedisian School of MedicineBostonMassachusettsUSA
| | - Sriram Ravindran
- Department of Oral Biology, College of DentistryUniversity of Illinois ChicagoChicagoIllinoisUSA
| | - Ashok K. Shetty
- Institute for Regenerative Medicine, Department of Cell Biology and GeneticsTexas A&M University School of MedicineCollege StationTexasUSA
| | - Johan Skog
- Exosome Diagnostics, a Bio‐Techne BrandWalthamMassachusettsUSA
| | - Kenneth W. Witwer
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Ali R. Djalilian
- Department of Ophthalmology and Visual SciencesUniversity of Illinois at ChicagoChicagoIllinoisUSA
| | - Alissa M. Weaver
- Department of Cell and Developmental BiologyVanderbilt University School of MedicineNashvilleTennesseeUSA
| |
Collapse
|