1
|
Tao K, Tao K, Wang J. The potential mechanisms of extracellular vesicles in transfusion-related adverse reactions: Recent advances. Transfus Clin Biol 2025:S1246-7820(25)00049-7. [PMID: 40180029 DOI: 10.1016/j.tracli.2025.03.006] [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: 01/15/2025] [Revised: 03/15/2025] [Accepted: 03/25/2025] [Indexed: 04/05/2025]
Abstract
Blood transfusion is an irreplaceable clinical treatment. Blood components are differentiated and stored according to specific guidelines. Storage temperatures and times vary depending on the blood component, but they all release extracellular vesicles (EVs) during storage. Although blood transfusions can be life-saving, they can also cause many adverse transfusion reactions, among which the effects of EVs are of increasing interest to researchers. EVs are submicron particles that vary in size, composition, and surface biomarkers, are encapsulated by a lipid bilayer, and are not capable of self-replication. EVs released by blood cells are important contributors to pathophysiologic states through proinflammatory, coagulant, and immunosuppressive effects, which in turn promote or inhibit the associated disease phenotype. Therefore, this review explores the potential mechanisms of hematopoietic-derived EVs in transfusion-associated adverse reactions and discusses the potential of the latest proteomics tools to be applied to the analysis of EVs in the field of transfusion medicine with a view to reducing the risk of blood transfusion.
Collapse
Affiliation(s)
- Keyi Tao
- Panzhihua University, Panzhihua 617000 Sichuan, China
| | - Keran Tao
- Institute of Medicine and Nursing, Hubei University of Medicine, Shiyan 442000 Hubei, China
| | - Jing Wang
- Southwest Medical University, Luzhou 646000 Sichuan, China; Department of Blood Transfusion, The Affiliated Hospital of Southwest Medical University, Luzhou Sichuan, 646000 China.
| |
Collapse
|
2
|
Chen J, Tan Q, Yang Z, Chen W, Zhou E, Li M, Deng J, Wu Y, Liu J, Xu J, Guo M, Jin Y. Dendritic Cell Derived-Extracellular Vesicles Engineered to Express Interleukin-12 and Anti-CTLA-4 on Their Surface for Combinational Cancer Immunotherapy. J Extracell Vesicles 2025; 14:e70068. [PMID: 40241233 PMCID: PMC12003100 DOI: 10.1002/jev2.70068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 03/11/2025] [Indexed: 04/18/2025] Open
Abstract
Dendritic cell (DC)-derived extracellular vesicles (DEVs) are promising candidates for cancer vaccines, but their therapeutic effects still need further optimization. In this study, we utilized neoantigens, lipopolysaccharide and IFN-γ to induce the maturation of DCs, and then isolated DEVs derived from these mature DCs. We showed that the immune checkpoint inhibitor (anti-CTLA-4 antibody, aCTLA-4) can improve the immunostimulatory function of DEVs by directly activating T cells through immune checkpoint signal blockade. The cytokine interleukin-12 (IL-12), as one of the third signals for T cell activation, can also enhance the capability of DEVs to activate T cells directly. Based on these findings, we designed the engineered DEVs conjugated with IL-12 and aCTLA-4 (DEV@IL-12-aCTLA-4) to improve the therapeutic potential of DEVs by providing sufficient immune regulatory signals. Moreover, the carrier property of DEVs also contributes to the delivery of IL-12 and aCTLA-4 to lymph nodes. This indicates that the conjugation of DEVs with IL-12 and aCTLA-4 constitutes a complementary approach, where IL-12 and aCTLA-4 help to enhance the T cell activation effect of DEVs, and DEVs facilitate the delivery of IL-12 and aCTLA-4. Our results showed that DEV@IL-12-aCTLA-4 can enhance the Th1 immune response and reverse exhausted CD8+ T cells in the tumour microenvironment, effectively inducing robust T cell immune responses and inhibiting tumour growth in tumour-bearing mice. Overall, this study expands the theoretical foundation of DEVs and provides a universal strategy for optimizing cancer combination immunotherapy by reprogramming DEVs.
Collapse
Affiliation(s)
- Jiangbin Chen
- Department of Respiratory and Critical Care Medicine, Hubei Province Clinical Research Center for Major Respiratory Diseases, and NHC Key Laboratory of Pulmonary DiseasesUnion Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanHubeiChina
- Hubei Province Key Laboratory of Biological Targeted Therapy, MOE Key Laboratory of Biological Targeted TherapyUnion Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanHubeiChina
- Hubei Province Engineering Research Center for Tumor‐Targeted BiochemotherapyUnion Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanHubeiChina
| | - Qi Tan
- Department of Respiratory and Critical Care Medicine, Hubei Province Clinical Research Center for Major Respiratory Diseases, and NHC Key Laboratory of Pulmonary DiseasesUnion Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanHubeiChina
- Hubei Province Key Laboratory of Biological Targeted Therapy, MOE Key Laboratory of Biological Targeted TherapyUnion Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanHubeiChina
- Hubei Province Engineering Research Center for Tumor‐Targeted BiochemotherapyUnion Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanHubeiChina
| | - Zimo Yang
- Department of Respiratory and Critical Care Medicine, Hubei Province Clinical Research Center for Major Respiratory Diseases, and NHC Key Laboratory of Pulmonary DiseasesUnion Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanHubeiChina
- Hubei Province Key Laboratory of Biological Targeted Therapy, MOE Key Laboratory of Biological Targeted TherapyUnion Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanHubeiChina
- Hubei Province Engineering Research Center for Tumor‐Targeted BiochemotherapyUnion Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanHubeiChina
| | - Wenjuan Chen
- Department of Respiratory and Critical Care Medicine, Hubei Province Clinical Research Center for Major Respiratory Diseases, and NHC Key Laboratory of Pulmonary DiseasesUnion Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanHubeiChina
- Hubei Province Key Laboratory of Biological Targeted Therapy, MOE Key Laboratory of Biological Targeted TherapyUnion Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanHubeiChina
- Hubei Province Engineering Research Center for Tumor‐Targeted BiochemotherapyUnion Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanHubeiChina
| | - E. Zhou
- Department of Respiratory and Critical Care Medicine, Hubei Province Clinical Research Center for Major Respiratory Diseases, and NHC Key Laboratory of Pulmonary DiseasesUnion Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanHubeiChina
- Hubei Province Key Laboratory of Biological Targeted Therapy, MOE Key Laboratory of Biological Targeted TherapyUnion Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanHubeiChina
- Hubei Province Engineering Research Center for Tumor‐Targeted BiochemotherapyUnion Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanHubeiChina
| | - Minglei Li
- Department of Respiratory and Critical Care Medicine, Hubei Province Clinical Research Center for Major Respiratory Diseases, and NHC Key Laboratory of Pulmonary DiseasesUnion Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanHubeiChina
- Hubei Province Key Laboratory of Biological Targeted Therapy, MOE Key Laboratory of Biological Targeted TherapyUnion Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanHubeiChina
- Hubei Province Engineering Research Center for Tumor‐Targeted BiochemotherapyUnion Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanHubeiChina
| | - Jingjing Deng
- Department of Respiratory and Critical Care Medicine, Hubei Province Clinical Research Center for Major Respiratory Diseases, and NHC Key Laboratory of Pulmonary DiseasesUnion Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanHubeiChina
- Hubei Province Key Laboratory of Biological Targeted Therapy, MOE Key Laboratory of Biological Targeted TherapyUnion Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanHubeiChina
- Hubei Province Engineering Research Center for Tumor‐Targeted BiochemotherapyUnion Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanHubeiChina
| | - Yali Wu
- Department of Respiratory and Critical Care Medicine, Hubei Province Clinical Research Center for Major Respiratory Diseases, and NHC Key Laboratory of Pulmonary DiseasesUnion Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanHubeiChina
- Hubei Province Key Laboratory of Biological Targeted Therapy, MOE Key Laboratory of Biological Targeted TherapyUnion Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanHubeiChina
- Hubei Province Engineering Research Center for Tumor‐Targeted BiochemotherapyUnion Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanHubeiChina
| | - Jiatong Liu
- Department of Respiratory and Critical Care Medicine, Hubei Province Clinical Research Center for Major Respiratory Diseases, and NHC Key Laboratory of Pulmonary DiseasesUnion Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanHubeiChina
- Hubei Province Key Laboratory of Biological Targeted Therapy, MOE Key Laboratory of Biological Targeted TherapyUnion Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanHubeiChina
- Hubei Province Engineering Research Center for Tumor‐Targeted BiochemotherapyUnion Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanHubeiChina
| | - Juanjuan Xu
- Department of Respiratory and Critical Care Medicine, Hubei Province Clinical Research Center for Major Respiratory Diseases, and NHC Key Laboratory of Pulmonary DiseasesUnion Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanHubeiChina
- Hubei Province Key Laboratory of Biological Targeted Therapy, MOE Key Laboratory of Biological Targeted TherapyUnion Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanHubeiChina
- Hubei Province Engineering Research Center for Tumor‐Targeted BiochemotherapyUnion Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanHubeiChina
| | - Mengfei Guo
- Department of Respiratory and Critical Care Medicine, Hubei Province Clinical Research Center for Major Respiratory Diseases, and NHC Key Laboratory of Pulmonary DiseasesUnion Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanHubeiChina
- Hubei Province Key Laboratory of Biological Targeted Therapy, MOE Key Laboratory of Biological Targeted TherapyUnion Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanHubeiChina
- Hubei Province Engineering Research Center for Tumor‐Targeted BiochemotherapyUnion Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanHubeiChina
| | - Yang Jin
- Department of Respiratory and Critical Care Medicine, Hubei Province Clinical Research Center for Major Respiratory Diseases, and NHC Key Laboratory of Pulmonary DiseasesUnion Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanHubeiChina
- Hubei Province Key Laboratory of Biological Targeted Therapy, MOE Key Laboratory of Biological Targeted TherapyUnion Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanHubeiChina
- Hubei Province Engineering Research Center for Tumor‐Targeted BiochemotherapyUnion Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanHubeiChina
| |
Collapse
|
3
|
Ten A, Yudintceva N, Samochernykh K, Combs SE, Jha HC, Gao H, Shevtsov M. Post-Secretion Processes and Modification of Extracellular Vesicles. Cells 2025; 14:408. [PMID: 40136657 PMCID: PMC11940929 DOI: 10.3390/cells14060408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 03/05/2025] [Accepted: 03/10/2025] [Indexed: 03/27/2025] Open
Abstract
Extracellular vesicles (EVs) are an important mediator of intercellular communication and the regulation of processes occurring in cells and tissues. The processes of EVs secretion by cells into the extracellular space (ECS) leads to their interaction with its participants. The ECS is a dynamic structure that also takes direct part in many processes of intercellular communication and regulation. Changes in the ECS can also be associated with pathological processes, such as increased acidity during the development of solid tumors, changes in the composition and nature of the organization of the extracellular matrix (ECM) during fibroblast activation, an increase in the content of soluble molecules during necrosis, and other processes. The interaction of these two systems, the EVs and the ESC, leads to structural and functional alteration in both participants. In the current review, we will focus on these alterations in the EVs which we termed post-secretory modification and processes (PSMPs) of EVs. PSPMs can have a significant effect on the immediate cellular environment and on the spread of the pathological process in the body as a whole. Thus, it can be assumed that PSPMs are one of the important stages in the regulation of intercellular communication, which has significant differences in the norm and in pathology.
Collapse
Affiliation(s)
- Artem Ten
- Laboratory of Biomedical Nanotechnologies, Institute of Cytology of the Russian Academy of Sciences (RAS), 194064 Saint Petersburg, Russia; (A.T.); (N.Y.)
| | - Natalia Yudintceva
- Laboratory of Biomedical Nanotechnologies, Institute of Cytology of the Russian Academy of Sciences (RAS), 194064 Saint Petersburg, Russia; (A.T.); (N.Y.)
- Personalized Medicine Centre, Almazov National Medical Research Centre, 2 Akkuratova Str., 197341 Saint Petersburg, Russia;
| | - Konstantin Samochernykh
- Personalized Medicine Centre, Almazov National Medical Research Centre, 2 Akkuratova Str., 197341 Saint Petersburg, Russia;
| | - Stephanie E. Combs
- Department of Radiation Oncology, Technishe Universität München (TUM), Klinikum Rechts der Isar, Ismaninger Str. 22, 81675 Munich, Germany;
| | - Hem Chandra Jha
- Department of Biosciences and Biomedical Engineering (BSBE), Indian Institute of Technology Indore, Khandwa Road, Simrol, Indore 453552, India;
| | - Huile Gao
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610064, China;
| | - Maxim Shevtsov
- Laboratory of Biomedical Nanotechnologies, Institute of Cytology of the Russian Academy of Sciences (RAS), 194064 Saint Petersburg, Russia; (A.T.); (N.Y.)
- Personalized Medicine Centre, Almazov National Medical Research Centre, 2 Akkuratova Str., 197341 Saint Petersburg, Russia;
- Department of Radiation Oncology, Technishe Universität München (TUM), Klinikum Rechts der Isar, Ismaninger Str. 22, 81675 Munich, Germany;
| |
Collapse
|
4
|
Wang J, Xing K, Zhang G, Li Z, Ding X, Leong DT. Surface Components and Biological Interactions of Extracellular Vesicles. ACS NANO 2025; 19:8433-8461. [PMID: 39999425 DOI: 10.1021/acsnano.4c16854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2025]
Abstract
Extracellular vesicles (EVs) are critical mediators of intercellular communication, carrying bioactive cargo and displaying diverse surface components that reflect their cellular origins and functions. The EV surface, composed of proteins, lipids, and glycocalyx elements, plays a pivotal role in targeting recipient cells, mediating biological interactions, and enabling selective cargo delivery. This review comprehensively examined the molecular architecture of EV surfaces, linking their biogenesis to functional diversity, and highlights their therapeutic and diagnostic potential in diseases such as cancer and cardiovascular disorders. Additionally, we explore emerging applications of EVs, including machine-learning-assisted analysis, chemical integration, and cross-system combinations. The review also discusses some key challenges in the clinical translation of EV-related technologies.
Collapse
Affiliation(s)
- Jinping Wang
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, 117585 Singapore
- School of Biological Science and Technology, University of Jinan, Jinan 250022, China
| | - Kuoran Xing
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, 117585 Singapore
| | - Guoying Zhang
- School of Biological Science and Technology, University of Jinan, Jinan 250022, China
| | - Zhiyang Li
- Clinical Laboratory, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing, Jiangsu Province 210008, China
| | - Xianguang Ding
- Key Laboratory for Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Nanjing University of Posts & Telecommunications, Nanjing 210023, China
| | - David Tai Leong
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, 117585 Singapore
| |
Collapse
|
5
|
Lin TI, Hsieh PY, Lin HJ, Chiang CK, Sheu JJC, Chang WT, Liau I, Hsu HY. Soy Protein-Cultured Mesenchymal Stem Cell-Secreted Extracellular Vesicles Target the Neurovascular Unit: Insights from a Zebrafish Brain Injury Model. ACS Biomater Sci Eng 2025; 11:1432-1444. [PMID: 40000145 PMCID: PMC11897944 DOI: 10.1021/acsbiomaterials.4c02304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 02/14/2025] [Accepted: 02/18/2025] [Indexed: 02/27/2025]
Abstract
Cerebral vascular disorders often accompany hypoxia-induced brain injury. In this study, we develop a zebrafish model of hypoxia-induced cerebral vascular injury to replicate the associated phenotypic changes, including cerebrovascular damage, neuronal apoptosis, and neurological dysfunction. We then explored the therapeutic potential of extracellular vesicles derived from Wharton's jelly-derived mesenchymal stem cells (WJ-MSCs) cultured on soy protein-coated surfaces. These vesicles demonstrated superior recovery efficacy, especially in restoring the blood-brain barrier integrity and improving neurological function. Our findings suggest that these potent therapeutic extracellular vesicles, easily produced from WJ-MSCs cultured in the presence of soy proteins, may mitigate hypoxia-induced brain injury by decreasing the severity of vascular disorder caused by oxidative stress. Protein-protein interactome analysis further suggests that multiple signaling pathways are likely involved in restoring normal neurovascular unit function.
Collapse
Affiliation(s)
- Tai-I Lin
- Department
of Applied Chemistry and Institute of Molecular Science, National Yang-Ming Chiao-Tung University, Hsinchu 300093, Taiwan
| | - Pei-Ying Hsieh
- Department
of Applied Chemistry and Institute of Molecular Science, National Yang-Ming Chiao-Tung University, Hsinchu 300093, Taiwan
| | - Hui-Jen Lin
- Department
of Applied Chemistry and Institute of Molecular Science, National Yang-Ming Chiao-Tung University, Hsinchu 300093, Taiwan
| | - Cheng-Kang Chiang
- Department
of Chemistry, National Dong Hwa University, Hualien 974301, Taiwan
| | - Jim Jinn-Chyuan Sheu
- Institute
of Biomedical Sciences, National Sun Yat-Sen
University, Kaohsiung 804201, Taiwan
| | - Wei-Tien Chang
- National
Taiwan University Hospital/National Taiwan University, Taipei 100233, Taiwan
| | - Ian Liau
- Department
of Applied Chemistry and Institute of Molecular Science, National Yang-Ming Chiao-Tung University, Hsinchu 300093, Taiwan
- Center
for Emergent Functional Matter Science, National Yang-Ming Chiao-Tung University, Hsinchu 300093, Taiwan
| | - Hsin-Yun Hsu
- Department
of Applied Chemistry and Institute of Molecular Science, National Yang-Ming Chiao-Tung University, Hsinchu 300093, Taiwan
- Center
for Emergent Functional Matter Science, National Yang-Ming Chiao-Tung University, Hsinchu 300093, Taiwan
| |
Collapse
|
6
|
Liu X, To KK, Zeng Q, Fu L. Effect of Extracellular Vesicles Derived From Tumor Cells on Immune Evasion. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2417357. [PMID: 39899680 PMCID: PMC11948033 DOI: 10.1002/advs.202417357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Indexed: 02/05/2025]
Abstract
The crosstalk between immunity and cancer in the regulation of tumor growth is considered a hallmark of cancer. Antitumor immunity refers to the innate and adaptive immune responses that regulate cancer development and proliferation. Tumor immune evasion represents a major hindrance to effective anticancer treatment. Extracellular vesicles (EVs) are nano-sized and lipid-bilayer-enclosed particles that are secreted to the extracellular space by all cell types. They are critically involved in numerous biological functions including intercellular communication. Tumor-derived extracellular vesicles (TEVs) can transport a variety of cargo to modulate immune cells in the tumor microenvironment (TME). This review provides the latest update about how tumor cells evade immune surveillance by exploiting TEVs. First, the biogenesis of EVs and the cargo-sorting machinery are discussed. Second, how tumor cells modulate immune cell differentiation, activation, and function via TEVs to evade immune surveillance is illustrated. Last but not least, the novel antitumor strategies that can reverse immune escape are summarized.
Collapse
Affiliation(s)
- Xuanfan Liu
- State Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for CancerCollaborative Innovation Center for Cancer MedicineGuangdong Esophageal Cancer InstituteSun Yat‐sen University Cancer CenterGuangzhou510060P. R. China
- Department of UrologyThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhou510080P. R. China
| | - Kenneth K.W. To
- School of PharmacyThe Chinese University of Hong KongHong Kong999077P. R. China
| | - Qinsong Zeng
- Department of UrologyThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhou510080P. R. China
- Guangxi Hospital Division of The First Affiliated HospitalSun Yat‐sen UniversityNanning530025P. R. China
| | - Liwu Fu
- State Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for CancerCollaborative Innovation Center for Cancer MedicineGuangdong Esophageal Cancer InstituteSun Yat‐sen University Cancer CenterGuangzhou510060P. R. China
| |
Collapse
|
7
|
Gurung S, Piskopos J, Steele J, Schittenhelm R, Shah A, Cousins FL, Tapmeier TT, Gargett CE. Potential Role of Menstrual Fluid-Derived Small Extracellular Vesicle Proteins in Endometriosis Pathogenesiss. J Extracell Vesicles 2025; 14:e70048. [PMID: 40091455 PMCID: PMC11911541 DOI: 10.1002/jev2.70048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 02/03/2025] [Indexed: 03/19/2025] Open
Abstract
Endometriosis, a chronic debilitating disease affects 1 in 7-10 girls and women, who have symptoms of severe chronic pain and subfertility and significantly impacts the overall quality of life. Currently, no effective early diagnostic methods are available for early stages of endometriosis. We used menstrual fluid-derived small extracellular vesicles (MF-sEVs) from women with self-reported endometriosis (laparoscopically diagnosed, n = 8) and self-reported without endometriosis and no painful periods (n = 9). MF-sEVs were separated using differential ultracentrifugation and characterised using nanoparticle tracking analysis (NTA), transmission electron microscopy (TEM), Western Blot, flow cytometry, mass-proteomics analysis and functional assays. Spherical-shaped sEVs were identified with a median diameter of ∼120 nm, expressing sEV marker proteins. The MF-sEV proteins were classified as endometrial origin. Over 5000 proteins were identified, ∼77% of which were decreased whilst only 22 proteins (largely comprising immunoglobulins) were increased in endometriosis/MF-sEVs compared to control/MF-sEVs. Decreased proteins were involved in nitrogen compound metabolism, immune response, intracellular signal transduction, regulation of programmed cell death, maintenance of cell polarity and actin cytoskeleton organisation. Flow cytometry demonstrated a significant increase in CD86 expression (immune activation marker) in endometriosis/MF-sEVs. Mesothelial cells showed a significant decrease in cellular resistance and junctional protein expression. MF-sEVs are possible contributors to the pathogenesis of endometriosis and may have the potential for early detection of the disease.
Collapse
Affiliation(s)
- Shanti Gurung
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Australia
- Obstetrics and Gynaecology, Monash University, Clayton, Australia
| | - Jacqueline Piskopos
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Australia
| | - Joel Steele
- Proteomics and Metabolomics Platform, Monash University, Clayton, Australia
| | - Ralf Schittenhelm
- Proteomics and Metabolomics Platform, Monash University, Clayton, Australia
| | - Anup Shah
- Proteomics and Metabolomics Platform, Monash University, Clayton, Australia
| | - Fiona L Cousins
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Australia
- Obstetrics and Gynaecology, Monash University, Clayton, Australia
| | - Thomas T Tapmeier
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Australia
- Obstetrics and Gynaecology, Monash University, Clayton, Australia
| | - Caroline E Gargett
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Australia
- Obstetrics and Gynaecology, Monash University, Clayton, Australia
| |
Collapse
|
8
|
Battaglini M, Carmignani A, Ciobanu DZ, Marino A, Catalano F, Armirotti A, Ciofani G. Detailed Profiling of Protein Corona Formed by Polydopamine Nanoparticles in Human Plasma. ACS APPLIED MATERIALS & INTERFACES 2025; 17:10485-10498. [PMID: 39909726 DOI: 10.1021/acsami.4c21207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2025]
Abstract
The term protein corona (PC) indicates proteins adsorbed onto the surface of nanostructures exposed to biological media such as blood or serum. The analysis of the composition, evolution, and effect of the PC complexed with nanomaterials gained attention in recent years due to the importance of these parameters in determining the biological fate of nanostructures. In particular, the PC represents the first component of a nanomaterial interfacing with biological structures, dictating parameters such as nanoparticle internalization, immune response, bioavailability, and even toxicity. Polydopamine nanoparticles (PDNPs), obtained through the polymerization of dopamine, are "smart" materials characterized by high biocompatibility, high antioxidant capacities, high tunability and surface reactivity, biodegradability, and the ability to act as photothermal conversion agents when irradiated with a near-infrared (NIR) light source. Despite many interesting applications of PDNPs are currently described in the scientific literature, there is still no comprehensive analysis of the phenomenon of PC formation consequent to the exposure of these nanomaterials to biological media. Moreover, to date, the investigation of the effects of light irradiation of photothermally active nanomaterials on the composition and evolution of the associated PC has been extremely limited. With this work, we aim to provide for the first time an analysis of the phenomenon of PC formation associated with PDNPs, before and after NIR light stimulation. We characterized the PC formed following exposure to human plasma and analyzed the effects of several parameters on the overall PC composition and quantity, such as the PDNP size, presence of a surface functionalization, exposure time, and irradiation with an NIR laser, demonstrating that these parameters play a pivotal role in the resulting PC composition. Eventually, we showed that PDNPs exposed to human plasma have significantly different properties with respect to bare PDNPs, showing higher internalization rates in human glioblastoma cells, a higher light absorption value, and enhanced photothermal conversion abilities.
Collapse
Affiliation(s)
- Matteo Battaglini
- Smart Bio-Interfaces, Istituto Italiano di Tecnologia, Viale Rinaldo Piaggio 34, Pontedera 56025, Italy
| | - Alessio Carmignani
- Smart Bio-Interfaces, Istituto Italiano di Tecnologia, Viale Rinaldo Piaggio 34, Pontedera 56025, Italy
| | - Dinu Zinovie Ciobanu
- Analytical Chemistry Facility, Istituto Italiano di Tecnologia, Via Morego 30, Genova 16163, Italy
| | - Attilio Marino
- Smart Bio-Interfaces, Istituto Italiano di Tecnologia, Viale Rinaldo Piaggio 34, Pontedera 56025, Italy
| | - Federico Catalano
- Electron Microscopy Facility, Istituto Italiano di Tecnologia, Via Morego 30, Genova 16163, Italy
| | - Andrea Armirotti
- Analytical Chemistry Facility, Istituto Italiano di Tecnologia, Via Morego 30, Genova 16163, Italy
| | - Gianni Ciofani
- Smart Bio-Interfaces, Istituto Italiano di Tecnologia, Viale Rinaldo Piaggio 34, Pontedera 56025, Italy
| |
Collapse
|
9
|
Pachane BC, Rodriguez BV, Shirk EN, Gololobova O, Carlson B, Queen SE, Erickson LD, Selistre-de-Araujo HS, Witwer KW. An ex vivo and in vitro investigation of extracellular vesicle interactions with B cells of Macaca nemestrina and humans. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.12.637883. [PMID: 39990430 PMCID: PMC11844526 DOI: 10.1101/2025.02.12.637883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
Extracellular vesicles may modify recipient cell behavior through multiple mechanisms, including interacting with the cell surface or internal membrane components and delivering luminal cargo to the cytoplasm. Here, we use a previously established ex vivo approach to investigate the cellular fate of EVs spiked into whole blood samples from nonhuman primate (NHP) and human donors and contrast these findings with results from in vitro assays. We report that EVs are internalized by NHP and human B cells while also associating to some degree with other PBMCs. EVs exhibit greater association with B cells in ex vivo whole blood compared to isolated B cells, suggesting that blood components may promote EV interactions or that cell isolation factors may inhibit this association. Cellular uptake of EVs involves clathrin-dependent endocytosis and may be aided by other pathways, including direct EV-cell membrane fusion. Overall, our data suggest that EV association, including uptake, by B cells occurs in at least two primate species. These findings highlight the potential to develop new strategies to either enhance or inhibit EV tropism toward B cells.
Collapse
|
10
|
Budayr OM, Miller BC, Nguyen J. Harnessing extracellular vesicle-mediated crosstalk between T cells and cancer cells for therapeutic applications. J Control Release 2025; 378:266-280. [PMID: 39657892 PMCID: PMC11830559 DOI: 10.1016/j.jconrel.2024.12.011] [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: 07/17/2024] [Revised: 11/23/2024] [Accepted: 12/05/2024] [Indexed: 12/12/2024]
Abstract
Small extracellular vesicles (EVs) are a diverse group of lipid-based particles that are ≤200 nm in diameter and contain an aqueous core. EVs have been shown to mediate intercellular communications between a wide array of immune cells; the downstream effects are diverse and have potential implications for the development of novel immunotherapeutic treatments. Despite a high volume of studies addressing the role EVs play in the immune system, our understanding of the crosstalk between T cells and cancer cells remains limited. Here, we discuss how EVs derived from cancer cells modulate T cell functions and conversely, how T cell derived EVs are crucial in modulating adaptive immune functions. In the context of cancer, tumor derived EVs (TD-EVs) halt T cell-mediated immunity by interfering with effector functions and enhancing regulatory T cell (Treg) functions. In contrast, EVs derived from effector T cells can serve to stimulate anticancer immunity, curbing metastasis and tumor growth. These findings highlight important aspects of how EVs can both mediate the therapeutic effects of T cells as well as impair T cell-mediated immunity. This calls for a deeper understanding of EV-mediated effects in order to advance them as next-generation therapeutics and nanocarriers.
Collapse
Affiliation(s)
- Omar M Budayr
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Brian C Miller
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Medicine, Division of Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27514, USA; Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27514, USA; Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| | - Juliane Nguyen
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
11
|
Yang Q, Failla AV, Turunen P, Mateos-Maroto A, Gai M, Zuschratter W, Westendorf S, Gelléri M, Chen Q, Goudappagouda, Zhao H, Zhu X, Morsbach S, Scheele M, Yan W, Landfester K, Kabe R, Bonn M, Narita A, Liu X. Reactivatable stimulated emission depletion microscopy using fluorescence-recoverable nanographene. Nat Commun 2025; 16:1341. [PMID: 39904997 PMCID: PMC11794581 DOI: 10.1038/s41467-025-56401-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 01/17/2025] [Indexed: 02/06/2025] Open
Abstract
Stimulated emission depletion (STED) microscopy, a key optical super-resolution imaging method, has extended our ability to view details to resolution levels of tens of nanometers. Its resolution depends on fluorophore de-excitation efficiency, and increases with depletion laser power. However, high-power irradiation permanently turns off the fluorescence due to photo-bleaching of the fluorophores. As a result, there is a trade-off between spatial resolution and imaging time. Here, we overcome this limitation by introducing reactivatable STED (ReSTED) based on the photophysical properties of the nanographene dibenzo[hi,st]ovalene (DBOV). In contrast to the photo-induced decomposition of other fluorophores, the fluorescence of DBOV is only temporarily deactivated and can be reactivated by near-infrared light (including the 775 nm depletion beam). As a result, this fluorophore allows for hours-long, high-resolution 3D STED imaging, greatly expanding the applications of STED microscopy.
Collapse
Affiliation(s)
- Qiqi Yang
- Max Planck Institute for Polymer Research, Mainz, Germany
| | - Antonio Virgilio Failla
- UKE Microscopy Imaging Facility, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Petri Turunen
- Institute of Molecular Biology gGmbH, Mainz, Germany
| | | | - Meiyu Gai
- Max Planck Institute for Polymer Research, Mainz, Germany
| | | | - Sophia Westendorf
- Institute of Physical and Theoretical Chemistry, University of Tuebingen, Tuebingen, Germany
| | | | - Qiang Chen
- Max Planck Institute for Polymer Research, Mainz, Germany
| | - Goudappagouda
- Organic and Carbon Nanomaterials Unit, Okinawa Institute of Science and Technology Graduate University, Onna-son, Okinawa, Japan
| | - Hao Zhao
- Organic and Carbon Nanomaterials Unit, Okinawa Institute of Science and Technology Graduate University, Onna-son, Okinawa, Japan
| | - Xingfu Zhu
- Max Planck Institute for Polymer Research, Mainz, Germany
| | | | - Marcus Scheele
- Institute of Physical and Theoretical Chemistry, University of Tuebingen, Tuebingen, Germany
| | - Wei Yan
- Key Laboratory of 3D Micro/Nano Fabrication and Characterization of Zhejiang Province, School of Engineering, Westlake University, Hangzhou, Zhejiang Province, China
| | | | - Ryota Kabe
- Organic Optoelectronics Unit, Okinawa Institute of Science and Technology Graduate University, Onna-son, Okinawa, Japan.
| | - Mischa Bonn
- Max Planck Institute for Polymer Research, Mainz, Germany.
| | - Akimitsu Narita
- Max Planck Institute for Polymer Research, Mainz, Germany.
- Organic and Carbon Nanomaterials Unit, Okinawa Institute of Science and Technology Graduate University, Onna-son, Okinawa, Japan.
| | - Xiaomin Liu
- Max Planck Institute for Polymer Research, Mainz, Germany.
| |
Collapse
|
12
|
Cominal JG, Gobbi Sebinelli H, Hayann L, Nogueira LFB, Cruz MAE, Mello MT, da Silva Andrilli LH, Bolean M, Ramos AP, Mebarek S, Bottini M, Millán JL, Ciancaglini P. A protein corona modulates the function of mineralization-competent matrix vesicles. JBMR Plus 2025; 9:ziae168. [PMID: 39877729 PMCID: PMC11772552 DOI: 10.1093/jbmrpl/ziae168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 12/03/2024] [Accepted: 12/19/2024] [Indexed: 01/31/2025] Open
Abstract
Mineralizing cells release a special class of extracellular vesicles known as matrix vesicles (MV), crucial for bone mineralization. Following their release, MV anchor to the extracellular matrix (ECM), where their highly specialized enzymatic machinery facilitates the formation of seed mineral within the MV's lumen, subsequently releasing it onto the ECM. However, how MV propagate mineral onto the collagenous ECM remains unclear. In this study, we address these questions by exploring the "protein corona" paradigm whereby nanoparticles entering a biological milieu become cloaked by a corona of soluble proteins modifying their biological functions. We isolated native MV from the growth plates of chicken embryos. After removing the protein corona from the native MV using high ionic strength buffer, we obtained shaved MV. Reconstituted MVs were produced by incubating shaved MV with the removed protein corona constituents. Our results show that both the removal and reconstitution of protein corona significantly affect the biochemical and physicochemical properties of MV, resulting in 3 well-defined groups. Shaved MV exhibited an increase in tissue nonspecific alkaline phosphatase (TNAP) activity and a decrease in mineral deposition compared to native MV. Reconstituted MV partially recovered these functions, showing a reduction of TNAP activity and mineral deposition compared to native MV. Furthermore, changes in the protein corona affect the MV ability to anchor to the collagenous ECM, which is crucial for initiating the propagation of the mineral phase within this organic matrix. Proteomic analyses revealed changes in the protein profile of the MV resulting from the removal of the protein corona, indicating that shaved proteins were primarily related to external structural and ECM organization and catabolism. These findings underscore the role of the protein corona in modulating the mineralization capabilities of MV. Understanding these interactions could lead to new therapeutic strategies for enhancing bone repair and regeneration.
Collapse
Affiliation(s)
- Juçara Gastaldi Cominal
- Departamento de Química, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto 14040-901, Brazil
| | - Heitor Gobbi Sebinelli
- Departamento de Química, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto 14040-901, Brazil
| | - Larwsk Hayann
- Departamento de Química, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto 14040-901, Brazil
| | - Lucas Fabrício Bahia Nogueira
- Departamento de Química, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto 14040-901, Brazil
| | - Marcos Antonio Eufrásio Cruz
- Departamento de Química, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto 14040-901, Brazil
| | - Maryanne Trafanni Mello
- Departamento de Química, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto 14040-901, Brazil
| | - Luiz Henrique da Silva Andrilli
- Departamento de Química, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto 14040-901, Brazil
- Sanford Children’s Health Research Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, United States
| | - Maytê Bolean
- Departamento de Química, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto 14040-901, Brazil
| | - Ana Paula Ramos
- Departamento de Química, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto 14040-901, Brazil
| | - Saida Mebarek
- Institut de Chimie et Biochimie Moléculaires et Supramoléculaires, UMR CNRS 5246, Université Claude Bernard Lyon 1, 69 622 Villeurbanne Cedex, France
| | - Massimo Bottini
- Sanford Children’s Health Research Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, United States
- Department of Experimental Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
| | - José Luis Millán
- Sanford Children’s Health Research Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, United States
| | - Pietro Ciancaglini
- Departamento de Química, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto 14040-901, Brazil
| |
Collapse
|
13
|
Musicò A, Zendrini A, Reyes SG, Mangolini V, Paolini L, Romano M, Papait A, Silini AR, Di Gianvincenzo P, Neva A, Cretich M, Parolini O, Almici C, Moya SE, Radeghieri A, Bergese P. Extracellular vesicles of different cellular origin feature distinct biomolecular corona dynamics. NANOSCALE HORIZONS 2024; 10:104-112. [PMID: 39559863 DOI: 10.1039/d4nh00320a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2024]
Abstract
Initially observed on synthetic nanoparticles, the existence of biomolecular corona and its role in determining nanoparticle identity and function are now beginning to be acknowledged in biogenic nanoparticles, particularly in extracellular vesicles - membrane-enclosed nanoparticle shuttling proteins, nucleic acids, and metabolites which are released by cells for physiological and pathological communication - we developed a methodology based on fluorescence correlation spectroscopy to track biomolecular corona formation on extracellular vesicles derived from human red blood cells and amniotic membrane mesenchymal stromal cells when these vesicles are dispersed in human plasma. The methodology allows for tracking corona dynamics in situ under physiological conditions. Results evidence that the two extracellular vesicle populations feature distinct corona dynamics. These findings indicate that the dynamics of the biomolecular corona may ultimately be linked to the cellular origin of the extracellular vesicles, revealing an additional level of heterogeneity, and possibly of bionanoscale identity, that characterizes circulating extracellular vesicles.
Collapse
Affiliation(s)
- Angelo Musicò
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy.
- CSGI, Center for Colloid and Surface Science, 50019 Florence, Italy
| | - Andrea Zendrini
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy.
- CSGI, Center for Colloid and Surface Science, 50019 Florence, Italy
| | - Santiago Gimenez Reyes
- Soft Matter Nanotechnology, Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), Paseo de Miramon 194, Donostia-San Sebastián, Spain
- Instituto de Fisica del Sur (IFISUR-CONICET), Av. Alem, Bahia Blanca, Argentina
| | - Valentina Mangolini
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy.
- CSGI, Center for Colloid and Surface Science, 50019 Florence, Italy
| | - Lucia Paolini
- CSGI, Center for Colloid and Surface Science, 50019 Florence, Italy
- Department of Medical and Surgical Specialties, Radiological Sciences and Public Health (DSMC), University of Brescia, Brescia, Italy
| | - Miriam Romano
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy.
- CSGI, Center for Colloid and Surface Science, 50019 Florence, Italy
| | - Andrea Papait
- Department of Life Science and Public Health, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
- Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, 00168 Rome, Italy
| | - Antonietta Rosa Silini
- Centro di Ricerca Eugenia Menni, Fondazione Poliambulanza Istituto Ospedaliero, 25124, Brescia, Italy
| | - Paolo Di Gianvincenzo
- Soft Matter Nanotechnology, Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), Paseo de Miramon 194, Donostia-San Sebastián, Spain
| | - Arabella Neva
- Laboratory for Stem Cells Manipulation and Cryopreservation, Department of Transfusion Medicine, ASST Spedali Civili of Brescia, 25123, Brescia, Italy
| | - Marina Cretich
- Istituto di Scienze e Tecnologie Chimiche "Giulio Natta" - National Research Council of Italy (SCITEC-CNR), 20131 Milan, Italy
| | - Ornella Parolini
- Department of Life Science and Public Health, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
- Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, 00168 Rome, Italy
| | - Camillo Almici
- Laboratory for Stem Cells Manipulation and Cryopreservation, Department of Transfusion Medicine, ASST Spedali Civili of Brescia, 25123, Brescia, Italy
| | - Sergio E Moya
- Soft Matter Nanotechnology, Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), Paseo de Miramon 194, Donostia-San Sebastián, Spain
| | - Annalisa Radeghieri
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy.
- CSGI, Center for Colloid and Surface Science, 50019 Florence, Italy
| | - Paolo Bergese
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy.
- CSGI, Center for Colloid and Surface Science, 50019 Florence, Italy
- National Inter-university Consortium of Materials Science and Technology (INSTM), Via Giuseppe Giusti 9, 50121 Firenze, Italy
| |
Collapse
|
14
|
Armengol-Badia O, Maggi J, Casal C, Cortés R, Abián J, Carrascal M, Closa D. The Microenvironment in an Experimental Model of Acute Pancreatitis Can Modify the Formation of the Protein Corona of sEVs, with Implications on Their Biological Function. Int J Mol Sci 2024; 25:12969. [PMID: 39684681 DOI: 10.3390/ijms252312969] [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: 10/04/2024] [Revised: 11/28/2024] [Accepted: 11/28/2024] [Indexed: 12/18/2024] Open
Abstract
A considerable number of the physiological functions of extracellular vesicles are conditioned by the protein corona attached to their surface. The composition of this corona is initially defined during their intracellular synthesis, but it can be subsequently modified by interactions with the microenvironment. Here, we evaluated how the corona of small extracellular vesicles exposed to the inflammatory environment generated in acute pancreatitis is modified and what functional changes occur as a result of these modifications. Small extracellular vesicles obtained from a pancreatic cell line were incubated with the ascitic fluid generated in experimental acute pancreatitis in rats. Using proteomic techniques, we detected the appearance of new proteins and an increase the uptake of extracellular vesicles by certain cell types and the response induced in inflammatory cells. The inhibition of different pattern recognition receptors reversed this activation, indicating that some of these effects could be due to binding of damage-associated molecular patterns to the corona. All of this indicates that in pathologies such as acute pancreatitis, characterized by an inflammatory response and intense tissue damage, the microenvironment substantially influences the corona of extracellular vesicles, thus altering their behavior and enhancing their inflammatory activity.
Collapse
Affiliation(s)
- Olga Armengol-Badia
- Department of Experimental Pathology, Institut d'Investigacions Biomèdiques de Barcelona, Consejo Superior de Investigaciones Científicas (IIBB-CSIC), Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
- Doctorate in Biotechnology, Facultat de Farmàcia i Ciències de l'Alimentació, Universitat de Barcelona, 08028 Barcelona, Spain
| | - Jaxaira Maggi
- Biological and Environmental Proteomics, Institut d'Investigacions Biomèdiques de Barcelona, Consejo Superior de Investigaciones Científicas (IIBB-CSIC), Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Carme Casal
- Microscopy Unit, Institut d'Investigacions Biomèdiques de Barcelona, Consejo Superior de Investigaciones Científicas (IIBB-CSIC), 08036 Barcelona, Spain
| | - Roser Cortés
- Department of Experimental Pathology, Institut d'Investigacions Biomèdiques de Barcelona, Consejo Superior de Investigaciones Científicas (IIBB-CSIC), Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Joaquín Abián
- Biological and Environmental Proteomics, Institut d'Investigacions Biomèdiques de Barcelona, Consejo Superior de Investigaciones Científicas (IIBB-CSIC), Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Montserrat Carrascal
- Biological and Environmental Proteomics, Institut d'Investigacions Biomèdiques de Barcelona, Consejo Superior de Investigaciones Científicas (IIBB-CSIC), Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Daniel Closa
- Department of Experimental Pathology, Institut d'Investigacions Biomèdiques de Barcelona, Consejo Superior de Investigaciones Científicas (IIBB-CSIC), Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| |
Collapse
|
15
|
Berger S, Zeyn Y, Wagner E, Bros M. New insights for the development of efficient DNA vaccines. Microb Biotechnol 2024; 17:e70053. [PMID: 39545748 PMCID: PMC11565620 DOI: 10.1111/1751-7915.70053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 10/29/2024] [Indexed: 11/17/2024] Open
Abstract
Despite the great potential of DNA vaccines for a broad range of applications, ranging from prevention of infections, over treatment of autoimmune and allergic diseases to cancer immunotherapies, the implementation of such therapies for clinical treatment is far behind the expectations up to now. The main reason is the poor immunogenicity of DNA vaccines in humans. Consequently, the improvement of the performance of DNA vaccines in vivo is required. This mini-review provides an overview of the current state of DNA vaccines and the various strategies to enhance the immunogenic potential of DNA vaccines, including (i) the optimization of the DNA construct itself regarding size, nuclear transfer and transcriptional regulation; (ii) the use of appropriate adjuvants; and (iii) improved delivery, for example, by careful choice of the administration route, physical methods such as electroporation and nanomaterials that may allow cell type-specific targeting. Moreover, combining nanoformulated DNA vaccines with other immunotherapies and prime-boost strategies may help to enhance success of treatment.
Collapse
Affiliation(s)
- Simone Berger
- Pharmaceutical Biotechnology, Department of Pharmacy, Center for NanoScienceLudwig‐Maximilians‐Universität (LMU) MunichMunichGermany
| | - Yanira Zeyn
- Department of DermatologyUniversity Medical Center of the Johannes Gutenberg University (JGU) MainzMainzGermany
| | - Ernst Wagner
- Pharmaceutical Biotechnology, Department of Pharmacy, Center for NanoScienceLudwig‐Maximilians‐Universität (LMU) MunichMunichGermany
| | - Matthias Bros
- Department of DermatologyUniversity Medical Center of the Johannes Gutenberg University (JGU) MainzMainzGermany
| |
Collapse
|
16
|
Jackson Cullison SR, Flemming JP, Karagoz K, Wermuth PJ, Mahoney MG. Mechanisms of extracellular vesicle uptake and implications for the design of cancer therapeutics. JOURNAL OF EXTRACELLULAR BIOLOGY 2024; 3:e70017. [PMID: 39483807 PMCID: PMC11522837 DOI: 10.1002/jex2.70017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 09/11/2024] [Accepted: 10/14/2024] [Indexed: 11/03/2024]
Abstract
The translation of pre-clinical anti-cancer therapies to regulatory approval has been promising, but slower than hoped. While innovative and effective treatments continue to achieve or seek approval, setbacks are often attributed to a lack of efficacy, failure to achieve clinical endpoints, and dose-limiting toxicities. Successful efforts have been characterized by the development of therapeutics designed to specifically deliver optimal and effective dosing to tumour cells while minimizing off-target toxicity. Much effort has been devoted to the rational design and application of synthetic nanoparticles to serve as targeted therapeutic delivery vehicles. Several challenges to the successful application of this modality as delivery vehicles include the induction of a protracted immune response that results in their rapid systemic clearance, manufacturing cost, lack of stability, and their biocompatibility. Extracellular vesicles (EVs) are a heterogeneous class of endogenous biologically produced lipid bilayer nanoparticles that mediate intercellular communication by carrying bioactive macromolecules capable of modifying cellular phenotypes to local and distant cells. By genetic, chemical, or metabolic methods, extracellular vesicles (EVs) can be engineered to display targeting moieties on their surface while transporting specific cargo to modulate pathological processes following uptake by target cell populations. This review will survey the types of EVs, their composition and cargoes, strategies employed to increase their targeting, uptake, and cargo release, and their potential as targeted anti-cancer therapeutic delivery vehicles.
Collapse
Affiliation(s)
| | - Joseph P. Flemming
- Rowan‐Virtua School of Osteopathic MedicineRowan UniversityStratfordNew JerseyUSA
| | - Kubra Karagoz
- Departments of PharmacologyPhysiology, and Cancer Biology, Thomas Jefferson UniversityPhiladelphiaPennsylvaniaUSA
| | | | - Mỹ G. Mahoney
- Departments of PharmacologyPhysiology, and Cancer Biology, Thomas Jefferson UniversityPhiladelphiaPennsylvaniaUSA
- Department of Otolaryngology – Head and Neck SurgeryThomas Jefferson UniversityPhiladelphiaPennsylvaniaUSA
| |
Collapse
|
17
|
Xia B, Hu R, Chen J, Shan S, Xu F, Zhang G, Zhou Z, Fan Y, Hu Z, Liang XJ. Oral Administration Properties Evaluation of Three Milk-Derived Extracellular Vesicles Based on Ultracentrifugation Extraction Methods. Adv Healthc Mater 2024; 13:e2401370. [PMID: 38767497 DOI: 10.1002/adhm.202401370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 05/16/2024] [Indexed: 05/22/2024]
Abstract
Milk-derived extracellular vesicles (M-EVs) are low-cost, can be prepared in large quantities, and can cross the gastrointestinal barrier for oral administration. However, the composition of milk is complex, and M-EVs obtained by different extraction methods may affect their oral delivery. Based on this, a new method for extracting M-EVs based on cryogenic freezing treatment (Cryo-M-EVs) is proposed and compared with the previously reported acetic acid treatment (Acid-M-EVs) method and the conventional ultracentrifugation method (Ulltr-M-EVs). The new method simplifies the pretreatment step and achieves 25-fold and twofold higher yields than Acid-M-EVs and Ulltr-M-EVs. And it is interesting to note that Cryo-M-EVs and Acid-M-EVs have higher cellular uptake efficiency, and Cryo-M-EVs present the best transepithelial transport effect. After oral administration of the three M-EVs extracted by three methods in mice, Cryo-M-EVs effectively successfully cross the gastrointestinal barrier and achieve hepatic accumulation, whereas Acid-M-EVs and Ultr-M-EVs mostly reside in the intestine. The M-EVs obtained by the three extraction methods show a favorable safety profile at the cellular as well as animal level. Therefore, when M-EVs obtained by different extraction methods are used for oral drug delivery, their accumulation properties at different sites can be utilized to better deal with different diseases.
Collapse
Affiliation(s)
- Bozhang Xia
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, No. 11, First North Road, Zhongguancun, Beijing, 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Runjing Hu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, No. 11, First North Road, Zhongguancun, Beijing, 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Junge Chen
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine & Shenzhen Institute of Beihang University, Beihang University, Beijing, 100083, China
| | - Shaobo Shan
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, No. 11, First North Road, Zhongguancun, Beijing, 100190, P. R. China
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100050, P. R. China
| | - Fengfei Xu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, No. 11, First North Road, Zhongguancun, Beijing, 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Gang Zhang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, No. 11, First North Road, Zhongguancun, Beijing, 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Ziran Zhou
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, No. 11, First North Road, Zhongguancun, Beijing, 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Yubo Fan
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine & Shenzhen Institute of Beihang University, Beihang University, Beijing, 100083, China
| | - Zhongbo Hu
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Xing-Jie Liang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, No. 11, First North Road, Zhongguancun, Beijing, 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| |
Collapse
|
18
|
Ye J, Li D, Jie Y, Luo H, Zhang W, Qiu C. Exosome-based nanoparticles and cancer immunotherapy. Biomed Pharmacother 2024; 179:117296. [PMID: 39167842 DOI: 10.1016/j.biopha.2024.117296] [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: 06/20/2024] [Revised: 08/06/2024] [Accepted: 08/09/2024] [Indexed: 08/23/2024] Open
Abstract
Over the past decades, cancer immunotherapy has encountered challenges such as immunogenicity, inefficiency, and cytotoxicity. Consequently, exosome-based cancer immunotherapy has gained rapid traction as a promising alternative. Exosomes, a type of extracellular vesicles (EVs) ranging from 50 to 150 nm, are self-originating and exhibit fewer side effects compared to traditional therapies. Exosome-based immunotherapy encompasses three significant areas: cancer vaccination, co-inhibitory checkpoints, and adoptive T-cell therapy. Each of these fields leverages the inherent advantages of exosomes, demonstrating substantial potential for individualized tumor therapy and precision medicine. This review aims to elucidate the reasons behind the promise of exosome-based nanoparticles as cancer therapies by examining their characteristics and summarizing the latest research advancements in cancer immunotherapy.
Collapse
Affiliation(s)
- Jiarong Ye
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang University, Jiangxi Province, 330000 China.
| | - Danni Li
- Second Clinical Medical School, Nanchang University, Jiangxi Province 330000, China
| | - Yiting Jie
- Second Clinical Medical School, Nanchang University, Jiangxi Province 330000, China
| | - Hongliang Luo
- Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, Jiangxi Province 330000, China
| | - Wenjun Zhang
- Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, Jiangxi Province 330000, China
| | - Cheng Qiu
- Gastrointestinal Surgery, Pingxiang People's Hospital, Jiangxi Province 330000, China.
| |
Collapse
|
19
|
Dlugolecka M, Czystowska-Kuzmicz M. Factors to consider before choosing EV labeling method for fluorescence-based techniques. Front Bioeng Biotechnol 2024; 12:1479516. [PMID: 39359260 PMCID: PMC11445045 DOI: 10.3389/fbioe.2024.1479516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 09/05/2024] [Indexed: 10/04/2024] Open
Abstract
A well-designed fluorescence-based analysis of extracellular vesicles (EV) can provide insights into the size, morphology, and biological function of EVs, which can be used in medical applications. Fluorescent nanoparticle tracking analysis with appropriate controls can provide reliable data for size and concentration measurements, while nanoscale flow cytometry is the most appropriate tool for characterizing molecular cargoes. Label selection is a crucial element in all fluorescence methods. The most comprehensive data can be obtained if several labeling approaches for a given marker are used, as they would provide complementary information about EV populations and interactions with the cells. In all EV-related experiments, the influence of lipoproteins and protein corona on the results should be considered. By reviewing and considering all the factors affecting EV labeling methods used in fluorescence-based techniques, we can assert that the data will provide as accurate as possible information about true EV biology and offer precise, clinically applicable information for future EV-based diagnostic or therapeutic applications.
Collapse
|
20
|
Xiao S, Wang J, Digiacomo L, Amici A, De Lorenzi V, Pugliese LA, Cardarelli F, Cerrato A, Laganà A, Cui L, Papi M, Caracciolo G, Marchini C, Pozzi D. Protein corona alleviates adverse biological effects of nanoplastics in breast cancer cells. NANOSCALE 2024; 16:16671-16683. [PMID: 39171675 DOI: 10.1039/d4nr01850h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/23/2024]
Abstract
Pollution from micro- and nanoplastics (MNPs) has long been a topic of concern due to its potential impact on human health. MNPs can circulate through human blood and, thus far, have been found in the lungs, spleen, stomach, liver, kidneys and even in the brain, placenta, and breast milk. While data are already available on the adverse biological effects of pristine MNPs (e.g. oxidative stress, inflammation, cytotoxicity, and even cancer induction), no report thus far clarified whether the same effects are modulated by the formation of a protein corona around MNPs. To this end, here we use pristine and human-plasma pre-coated polystyrene (PS) nanoparticles (NPs) and investigate them in cultured breast cancer cells both in terms of internalization and cell biochemical response to the exposure. It is found that pristine NPs tend to stick to the cell membrane and inhibit HER-2-driven signaling pathways, including phosphatidylinositol-3-kinase (PI3K)/protein kinase B (AKT) and mitogen-activated protein kinase (MAPK)/extracellular signal-regulated kinase (ERK) pathways, which are associated with cancer cell survival and growth. By contrast, the formation of a protein corona around the same NPs can promote their uptake by endocytic vesicles and final sequestration within lysosomes. Of note is that such intracellular fate of PS-NPs is associated with mitigation of the biochemical alterations of the phosphorylated AKT (pAKT)/AKT and phosphorylated ERK (pERK)/ERK levels. These findings provide the distribution of NPs in human breast cancer cells, may broaden our understanding of the interactions between NPs and breast cancer cells and underscore the crucial role of the protein corona in modulating the impact of MNPs on human health.
Collapse
Affiliation(s)
- Siyao Xiao
- NanoDelivery Lab, Department of Molecular Medicine, Sapienza University of Rome, Viale Regina Elena 291, 00161 Rome, Italy.
| | - Junbiao Wang
- School of Biosciences and Veterinary Medicine, University of Camerino, 62032 Camerino, Italy.
| | - Luca Digiacomo
- NanoDelivery Lab, Department of Molecular Medicine, Sapienza University of Rome, Viale Regina Elena 291, 00161 Rome, Italy.
| | - Augusto Amici
- School of Biosciences and Veterinary Medicine, University of Camerino, 62032 Camerino, Italy.
| | - Valentina De Lorenzi
- Laboratorio NEST, Scuola Normale Superiore, Piazza San Silvestro 12, 56127 Pisa, Italy
| | - Licia Anna Pugliese
- Laboratorio NEST, Scuola Normale Superiore, Piazza San Silvestro 12, 56127 Pisa, Italy
| | - Francesco Cardarelli
- Laboratorio NEST, Scuola Normale Superiore, Piazza San Silvestro 12, 56127 Pisa, Italy
| | - Andrea Cerrato
- Department of Chemistry, Sapienza University of Rome, P.le A. Moro 5, 00185 Rome, Italy
| | - Aldo Laganà
- Department of Chemistry, Sapienza University of Rome, P.le A. Moro 5, 00185 Rome, Italy
| | - Lishan Cui
- Department of Neuroscience, Catholic University of the Sacred Heart, Largo Francesco Vito 1, Rome 00168, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCSS, Rome 00168, Italy
| | - Massimiliano Papi
- Department of Neuroscience, Catholic University of the Sacred Heart, Largo Francesco Vito 1, Rome 00168, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCSS, Rome 00168, Italy
| | - Giulio Caracciolo
- NanoDelivery Lab, Department of Molecular Medicine, Sapienza University of Rome, Viale Regina Elena 291, 00161 Rome, Italy.
| | - Cristina Marchini
- School of Biosciences and Veterinary Medicine, University of Camerino, 62032 Camerino, Italy.
| | - Daniela Pozzi
- NanoDelivery Lab, Department of Molecular Medicine, Sapienza University of Rome, Viale Regina Elena 291, 00161 Rome, Italy.
| |
Collapse
|
21
|
Zhao J, Zhu W, Mao Y, Li X, Ling G, Luo C, Zhang P. Unignored intracellular journey and biomedical applications of extracellular vesicles. Adv Drug Deliv Rev 2024; 212:115388. [PMID: 38969268 DOI: 10.1016/j.addr.2024.115388] [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: 04/13/2024] [Revised: 06/02/2024] [Accepted: 07/01/2024] [Indexed: 07/07/2024]
Abstract
The intracellular journey of extracellular vesicles (EVs) cannot be ignored in various biological pathological processes. In this review, the biogenesis, biological functions, uptake pathways, intracellular trafficking routes, and biomedical applications of EVs were highlighted. Endosomal escape is a unique mode of EVs release. When vesicles escape from endosomes, they avoid the fate of fusing with lysosomes and being degraded, thus having the opportunity to directly enter the cytoplasm or other organelles. This escape mechanism is crucial for EVs to deliver specific signals or substances. The intracellular trafficking of EVs after endosomal escape is a complex and significant biological process that involves the coordinated work of various cellular structures and molecules. Through the in-depth study of this process, the function and regulatory mechanism of EVs are fully understood, providing new dimensions for future biomedical diagnosis and treatment.
Collapse
Affiliation(s)
- Jiuhong Zhao
- Wuya College of Innovation, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, China
| | - Wenjing Zhu
- Wuya College of Innovation, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, China
| | - Yuxuan Mao
- Wuya College of Innovation, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, China
| | - Xiaodan Li
- Wuya College of Innovation, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, China
| | - Guixia Ling
- Wuya College of Innovation, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, China.
| | - Cong Luo
- Wuya College of Innovation, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, China; Joint International Research Laboratory of Intelligent Drug Delivery Systems of Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China.
| | - Peng Zhang
- Wuya College of Innovation, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, China.
| |
Collapse
|
22
|
Spokeviciute B, Kholia S, Brizzi MF. Chimeric antigen receptor (CAR) T-cell therapy: Harnessing extracellular vesicles for enhanced efficacy. Pharmacol Res 2024; 208:107352. [PMID: 39147005 DOI: 10.1016/j.phrs.2024.107352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 08/05/2024] [Accepted: 08/12/2024] [Indexed: 08/17/2024]
Abstract
A cutting-edge approach in cell-based immunotherapy for combating resistant cancer involves genetically engineered chimeric antigen receptor T (CAR-T) lymphocytes. In recent years, these therapies have demonstrated effectiveness, leading to their commercialization and clinical application against certain types of cancer. However, CAR-T therapy faces limitations, such as the immunosuppressive tumour microenvironment (TME) that can render CAR-T cells ineffective, and the adverse side effects of the therapy, including cytokine release syndrome (CRS). Extracellular vesicles (EVs) are a diverse group of membrane-bound particles released into the extracellular environment by virtually all cell types. They are essential for intercellular communication, transferring cargoes such as proteins, lipids, various types of RNAs, and DNA fragments to target cells, traversing biological barriers both locally and systemically. EVs play roles in numerous physiological processes, with those from both immune and non-immune cells capable of modulating the immune system through activation or suppression. Leveraging this capability of EVs to enhance CAR-T cell therapy could represent a significant advancement in overcoming its current limitations. This review examines the current landscape of CAR-T cell immunotherapy and explores the potential role of EVs in augmenting its therapeutic efficacy.
Collapse
Affiliation(s)
| | - Sharad Kholia
- Department of Medical Sciences, University of Torino, Turin, Italy
| | | |
Collapse
|
23
|
Caracciolo G. Artificial protein coronas: directing nanoparticles to targets. Trends Pharmacol Sci 2024; 45:602-613. [PMID: 38811308 DOI: 10.1016/j.tips.2024.05.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Revised: 04/25/2024] [Accepted: 05/09/2024] [Indexed: 05/31/2024]
Abstract
The protein corona surrounding nanoparticles (NPs) offers exciting possibilities for targeted drug delivery. However, realizing this potential requires direct evidence of corona-receptor interactions in vivo; a challenge hampered by the limitations of in vitro settings. This opinion proposes that utilizing engineered protein coronas can address this challenge. Artificial coronas made of selected plasma proteins retain their properties in vivo, enabling manipulation for specific receptor targeting. To directly assess corona-receptor interactions mimicking in vivo complexity, we propose testing artificial coronas with recently adapted quartz crystal microbalance (QCM) setups whose current limitations and potential advancements are critically discussed. Finally, the opinion proposes future experiments to decipher corona-receptor interactions and unlock the full potential of the protein corona for NP-based drug delivery.
Collapse
Affiliation(s)
- Giulio Caracciolo
- NanoDelivery Lab, Department of Molecular Medicine, Sapienza University of Rome, V.le Regina Elena 291, 00161, Rome, Italy.
| |
Collapse
|
24
|
Sharma A, Yadav A, Nandy A, Ghatak S. Insight into the Functional Dynamics and Challenges of Exosomes in Pharmaceutical Innovation and Precision Medicine. Pharmaceutics 2024; 16:709. [PMID: 38931833 PMCID: PMC11206934 DOI: 10.3390/pharmaceutics16060709] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Revised: 05/13/2024] [Accepted: 05/16/2024] [Indexed: 06/28/2024] Open
Abstract
Of all the numerous nanosized extracellular vesicles released by a cell, the endosomal-originated exosomes are increasingly recognized as potential therapeutics, owing to their inherent stability, low immunogenicity, and targeted delivery capabilities. This review critically evaluates the transformative potential of exosome-based modalities across pharmaceutical and precision medicine landscapes. Because of their precise targeted biomolecular cargo delivery, exosomes are posited as ideal candidates in drug delivery, enhancing regenerative medicine strategies, and advancing diagnostic technologies. Despite the significant market growth projections of exosome therapy, its utilization is encumbered by substantial scientific and regulatory challenges. These include the lack of universally accepted protocols for exosome isolation and the complexities associated with navigating the regulatory environment, particularly the guidelines set forth by the U.S. Food and Drug Administration (FDA). This review presents a comprehensive overview of current research trajectories aimed at addressing these impediments and discusses prospective advancements that could substantiate the clinical translation of exosomal therapies. By providing a comprehensive analysis of both the capabilities and hurdles inherent to exosome therapeutic applications, this article aims to inform and direct future research paradigms, thereby fostering the integration of exosomal systems into mainstream clinical practice.
Collapse
Affiliation(s)
| | | | | | - Subhadip Ghatak
- McGowan Institute for Regenerative Medicine, Department of Surgery, University of Pittsburgh, Pittsburgh, PA 15219, USA; (A.S.); (A.Y.); (A.N.)
| |
Collapse
|