1
|
Lim Falk V, Mueller-Wirth N, Karathanasis D, Evangelopoulos ME, Maleska Maceski A, Zadic A, Kuhle J, Schlup C, Marti S, Guse K, Chan A, Pernet V. Extracellular Vesicle Marker Changes Associated With Disease Activity in Relapsing-Remitting Multiple Sclerosis. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2025; 12:e200404. [PMID: 40300121 PMCID: PMC12056760 DOI: 10.1212/nxi.0000000000200404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 03/19/2025] [Indexed: 05/01/2025]
Abstract
BACKGROUND AND OBJECTIVES Multiple sclerosis (MS), neuromyelitis optica spectrum disorder (NMOSD), and myelin oligodendrocyte glycoprotein antibody-associated disease (MOGAD) are autoimmune disorders of the CNS causing severe neurologic impairment. Evidence suggests that extracellular vesicles (EVs) may play a disease-specific role in the orchestration of the immune cell response of MS, NMOSD, and MOGAD. In addition, EVs are considered as a potential source of biomarkers that may allow us to establish molecular signatures for these diseases and perhaps as well to follow treatment effects and disease progression. The aim of this study was to analyze the composition of EVs in patients with relapsing-remitting MS (RRMS) (n = 52), NMOSD (n = 19), and MOGAD (n = 10) and healthy controls ([HCs], n = 15). METHODS The concentrations of neurofilament light chain (NfL) and glial fibrillary acidic protein (GFAP) were determined in plasma using single-molecule array (SIMOA). The size and concentration of tetraspanin-presenting EVs were evaluated in plasma samples with a single-particle interferometric resonance imaging sensor (SP-IRIS). Tetraspanin-independent analyses were performed by nanoparticle-tracking analysis (NTA) after EV isolation by size exclusion (SmartSEC) and cryo-electron microscopy observations. EV epitopes were analyzed by extended multiplex analysis using flow cytometry. RESULTS The plasma concentration of NfL and GFAP was significantly higher in patients with RRMS than in HCs. For patients with NMOSD, only GFAP increased. The density of EVs assessed by NTA was lower in plasma of patients with RRMS than in HC plasma. In addition, the 3 disease groups presented increased mean EV sizes in comparison with HCs. Tetraspanin-based EV analyses by SP-IRIS allowed us to observe a modest difference in the level of CD81 in RRMS EVs. In patients with RRMS, but not in those with NMOSD and MOGAD, multiplex/flow cytometry analyses revealed changes in the EV levels of CD29, CD31, and CD69 associated with the time elapsed since the last relapse. The negative correlations established between the vesicular levels of CD31, CD40, CD44, CD49c, CD69, and NfL or GFAP z-scores suggest a negative relationship specifically in RRMS. DISCUSSION We speculate that the higher release of EVs containing CD29, CD31, CD40, CD44, CD49c, and CD69 in plasma, at low levels of circulating NfL/GFAP, may be associated with reduced immune cell activity in RRMS. These EV markers may characterize patients with RRMS in disease stabilization.
Collapse
Affiliation(s)
- Victoria Lim Falk
- Department of Neurology, Inselspital, Bern University Hospital, University of Bern, Switzerland
- Department of BioMedical Research (DBMR), University of Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences (GCB), University of Bern, Switzerland
| | - Nicole Mueller-Wirth
- CSL Behring, CSL Biologics Research Center, Bern, Switzerland
- Swiss Institute for Translational and Entrepreneurial Medicine, Sitem-Insel, Bern, Switzerland
| | | | | | - Aleksandra Maleska Maceski
- Departments of Biomedicine and Clinical Research, Multiple Sclerosis Centre and Research Center for Clinical Neuroimmunology and Neuroscience (RC2NB), University Hospital and University of Basel, Switzerland
- Department of Neurology, University Hospital and University of Basel, Switzerland
| | - Amar Zadic
- Departments of Biomedicine and Clinical Research, Multiple Sclerosis Centre and Research Center for Clinical Neuroimmunology and Neuroscience (RC2NB), University Hospital and University of Basel, Switzerland
- Department of Neurology, University Hospital and University of Basel, Switzerland
| | - Jens Kuhle
- Departments of Biomedicine and Clinical Research, Multiple Sclerosis Centre and Research Center for Clinical Neuroimmunology and Neuroscience (RC2NB), University Hospital and University of Basel, Switzerland
- Department of Neurology, University Hospital and University of Basel, Switzerland
| | - Cornelia Schlup
- CSL Behring, CSL Biologics Research Center, Bern, Switzerland
- Swiss Institute for Translational and Entrepreneurial Medicine, Sitem-Insel, Bern, Switzerland
| | - Stefanie Marti
- Department of Neurology, Inselspital, Bern University Hospital, University of Bern, Switzerland
- Department of BioMedical Research (DBMR), University of Bern, Switzerland
| | - Kirsten Guse
- CSL Behring, CSL Biologics Research Center, Bern, Switzerland
- Swiss Institute for Translational and Entrepreneurial Medicine, Sitem-Insel, Bern, Switzerland
| | - Andrew Chan
- Department of Neurology, Inselspital, Bern University Hospital, University of Bern, Switzerland
- Department of BioMedical Research (DBMR), University of Bern, Switzerland
| | - Vincent Pernet
- Department of Neurology, Inselspital, Bern University Hospital, University of Bern, Switzerland
- Department of BioMedical Research (DBMR), University of Bern, Switzerland
- Regenerative Medicine Unit, University Hospital Center of Quebec, Laval University, Quebec City, Canada; and
- Department of Molecular Medicine, Faculty of Medicine, Laval University, Quebec City, Canada
| |
Collapse
|
2
|
Ahn W, Han J, Kim N, Hwang YH, Kim W, Lee Y, Lee DY, Cheong IW, Han K, Nam GH, Kim IS, Lee EJ. Hierarchical protein nano-crystalline hydrogel with extracellular vesicles for ectopic lymphoid structure formation. Biomaterials 2025; 318:123166. [PMID: 39933315 DOI: 10.1016/j.biomaterials.2025.123166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 01/21/2025] [Accepted: 02/03/2025] [Indexed: 02/13/2025]
Abstract
Among cancer therapies, immune checkpoint blockade (ICB) has emerged as a prominent approach, substantially enhancing anti-tumor immune responses. However, the efficacy of ICB is often limited in the absence of a pre-existing immune response within the tumor microenvironment. Here, we introduce a novel hierarchical protein hydrogel platform designed to facilitate the formation of artificial tertiary lymphoid structures (aTLS), thereby improving ICB efficacy. Through the integration of self-assembling ferritin protein nanocages, rec1-resilin protein, and CP05 peptide, our hierarchical hydrogels provide a structurally supportive and functionally adaptive scaffold capable of on-demand self-repair in response to mild thermal treatments. The effective encapsulation of extracellular vesicles (EVs) via the CP05 peptide ensures the formation of aTLS with germinal center-like structures within the hierarchical hydrogel. We demonstrate that, combined with ICB therapy, EV-loaded hierarchical hydrogels also induce the TLS within the tumor, markedly promoting immune responses against ICB-resistant tumor. This bioactive hydrogel platform offers a versatile tool for enhancing a broad range of immunotherapies, with potential applications extending beyond TLS to other frameworks that support complex tissue architectures.
Collapse
Affiliation(s)
- Wonkyung Ahn
- Department of Chemical Engineering, School of Chemical Engineering and Applied Chemistry, Kyungpook National University, Daegu, 41566, Republic of Korea; Department of Biochemistry & Molecular Biology, Korea University College of Medicine, Seoul, 02841, Republic of Korea
| | - Jihoon Han
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02842, Republic of Korea; Chemical & Biological Integrative Research Center, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
| | - Nayeon Kim
- Department of Chemical Engineering, School of Chemical Engineering and Applied Chemistry, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Yeong Ha Hwang
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02842, Republic of Korea; Chemical & Biological Integrative Research Center, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
| | - Wonjun Kim
- Department of Chemical Engineering, School of Chemical Engineering and Applied Chemistry, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Yeram Lee
- Department of Chemical Engineering, School of Chemical Engineering and Applied Chemistry, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Dong Yun Lee
- Department of Polymer Science and Engineering, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - In Woo Cheong
- Department of Applied Chemistry, School of Chemical Engineering and Applied Chemistry, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Koohee Han
- Department of Chemical Engineering, School of Chemical Engineering and Applied Chemistry, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Gi-Hoon Nam
- Department of Biochemistry & Molecular Biology, Korea University College of Medicine, Seoul, 02841, Republic of Korea
| | - In-San Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02842, Republic of Korea; Chemical & Biological Integrative Research Center, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea.
| | - Eun Jung Lee
- Department of Chemical Engineering, School of Chemical Engineering and Applied Chemistry, Kyungpook National University, Daegu, 41566, Republic of Korea.
| |
Collapse
|
3
|
Trifylli EM, Angelakis A, Kriebardis AG, Papadopoulos N, Fortis SP, Pantazatou V, Koskinas J, Kranidioti H, Koustas E, Sarantis P, Manolakopoulos S, Deutsch M. Extracellular vesicles as biomarkers for metabolic dysfunction-associated steatotic liver disease staging using explainable artificial intelligence. World J Gastroenterol 2025; 31:106937. [DOI: 10.3748/wjg.v31.i22.106937] [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: 03/10/2025] [Revised: 04/18/2025] [Accepted: 05/22/2025] [Indexed: 06/12/2025] Open
Abstract
BACKGROUND Metabolic dysfunction-associated steatotic liver disease (MASLD) is a leading cause of chronic liver disease globally. Current diagnostic methods, such as liver biopsies, are invasive and have limitations, highlighting the need for non-invasive alternatives.
AIM To investigate extracellular vesicles (EVs) as potential biomarkers for diagnosing and staging steatosis in patients with MASLD using machine learning (ML) and explainable artificial intelligence (XAI).
METHODS In this single-center observational study, 798 patients with metabolic dysfunction were enrolled. Of these, 194 met the eligibility criteria, and 76 successfully completed all study procedures. Transient elastography was used for steatosis and fibrosis staging, and circulating plasma EV characteristics were analyzed through nanoparticle tracking. Twenty ML models were developed: Six to differentiate non-steatosis (S0) from steatosis (S1-S3); and fourteen to identify severe steatosis (S3). Models utilized EV features (size and concentration), clinical (advanced fibrosis and presence of type 2 diabetes mellitus), and anthropomorphic (sex, age, height, weight, body mass index) data. Their performance was assessed using receiver operating characteristic (ROC)-area under the curve (AUC), specificity, and sensitivity, while correlation and XAI analysis were also conducted.
RESULTS The CatBoost C1a model achieved an ROC-AUC of 0.71/0.86 (train/test) on average across ten random five-fold cross-validations, using EV features alone to distinguish S0 from S1-S3. The CatBoost C2h-21 model achieved an ROC-AUC of 0.81/1.00 (train/test) on average across ten random three-fold cross-validations, using engineered features including EVs, clinical features like diabetes and advanced fibrosis, and anthropomorphic data like body mass index and weight for identifying severe steatosis (S3). Key predictors included EV mean size and concentration. Correlation, XAI, and SHapley Additive exPlanations analysis revealed non-linear feature relationships with steatosis stages.
CONCLUSION The EV-based ML models demonstrated that the mean size and concentration of circulating plasma EVs constituted key predictors for distinguishing the absence of significant steatosis (S0) in patients with metabolic dysfunction, while the combination of EV, clinical, and anthropomorphic features improved the diagnostic accuracy for the identification of severe steatosis. The algorithmic approach using ML and XAI captured non-linear patterns between disease features and provided interpretable MASLD staging insights. However, further large multicenter studies, comparisons, and validation with histopathology and advanced imaging methods are needed.
Collapse
Affiliation(s)
- Eleni Myrto Trifylli
- Gastrointestinal-Liver Unit, The 2nd Department of Internal Medicine, National and Kapodistrian University of Athens, General Hospital of Athens “Hippocratio,” Athens 11521, Greece
- Laboratory of Reliability and Quality Control in Laboratory Hematology, Department of Biomedical Sciences, Section of Medical Laboratories, School of Health & Caring Sciences, University of West Attica, Egaleo 12243, Attikí, Greece
| | - Athanasios Angelakis
- Department of Epidemiology and Data Science, Amsterdam University Medical Center, Amsterdam 1105, Netherlands
- Department of Methodology, Digital Health, Amsterdam Public Health Research Institute, Amsterdam 1105, Netherlands
- Data Science Center, University of Amsterdam, Amsterdam 1098, Netherlands
| | - Anastasios G Kriebardis
- Laboratory of Reliability and Quality Control in Laboratory Hematology, Department of Biomedical Sciences, Section of Medical Laboratories, School of Health & Caring Sciences, University of West Attica, Egaleo 12243, Attikí, Greece
| | - Nikolaos Papadopoulos
- The Second Department of Internal Medicine, 401 General Army Hospital of Athens, Athens 11525, Greece
| | - Sotirios P Fortis
- Laboratory of Reliability and Quality Control in Laboratory Hematology, Department of Biomedical Sciences, Section of Medical Laboratories, School of Health & Caring Sciences, University of West Attica, Egaleo 12243, Attikí, Greece
| | - Vasiliki Pantazatou
- Laboratory of Reliability and Quality Control in Laboratory Hematology, Department of Biomedical Sciences, Section of Medical Laboratories, School of Health & Caring Sciences, University of West Attica, Egaleo 12243, Attikí, Greece
| | - John Koskinas
- Gastrointestinal-Liver Unit, The 2nd Department of Internal Medicine, National and Kapodistrian University of Athens, General Hospital of Athens “Hippocratio,” Athens 11521, Greece
| | - Hariklia Kranidioti
- Gastrointestinal-Liver Unit, The 2nd Department of Internal Medicine, National and Kapodistrian University of Athens, General Hospital of Athens “Hippocratio,” Athens 11521, Greece
| | - Evangelos Koustas
- Department of Oncology, General Hospital Evangelismos, Athens 10676, Greece
| | - Panagiotis Sarantis
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Athens 11527, Greece
| | - Spilios Manolakopoulos
- Gastrointestinal-Liver Unit, The 2nd Department of Internal Medicine, National and Kapodistrian University of Athens, General Hospital of Athens “Hippocratio,” Athens 11521, Greece
| | - Melanie Deutsch
- Gastrointestinal-Liver Unit, The 2nd Department of Internal Medicine, National and Kapodistrian University of Athens, General Hospital of Athens “Hippocratio,” Athens 11521, Greece
| |
Collapse
|
4
|
Ding L, Guo Q, Ren Y, Wang P, Pan Y, Ding J, Wang F, Yang Z, Zhu H. Construction of [ 89Zr]Zr-Labeled Human Umbilical Cord Mesenchymal Stem Cell-Derived Extracellular Vesicles for Noninvasive Detection of Tumors. ACS APPLIED MATERIALS & INTERFACES 2025. [PMID: 40387191 DOI: 10.1021/acsami.5c03280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2025]
Abstract
Extracellular vesicles (EVs) act as carriers that transfer molecules between donor and recipient cells, thereby altering the phenotype and function of the latter. EVs derived from mesenchymal stem cells (MSCs) inherit the homing ability of their parent cells to tumor sites. We speculate that MSC-derived EVs labeled with the isotope zirconium-89 ([89Zr]Zr) will accumulate within tumors and have the potential for tumor location via positron emission tomography (PET) imaging. Here, as a proof of concept, we used [89Zr]Zr-labeled human umbilical cord MSC (hucMSC)-derived EVs to characterize the homing of EVs to tumor regions via PET imaging. The uptake of [89Zr]Zr-DFO-hucMSC-EVs by 4T1, H292, and FaDu cells increased in a time-dependent manner. [89Zr]Zr-DFO-hucMSC-EVs were rapidly cleared from the blood circulation, showing a 2-phase exponential decay with a biodistribution half-life of 0.46 h and an elimination-phase half-life of 11.87 h. At 24 h postinjection, [89Zr]Zr-DFO-hucMSC-EVs were mainly distributed in the liver (10.39 ± 0.52%ID/g) and, to a lesser extent, in the spleen (9.87 ± 0.87%ID/g). PET imaging in tumor-bearing mouse models revealed persistently high tumor accumulation. The projected effective dose for an adult human female was low and was 0.084 mSv/MBq. There was no obvious normal tissue toxicity following the administration of excessive radioactivity. In summary, these studies demonstrate the potential feasibility of using the [89Zr]Zr-DFO-hucMSC-EV tracer for noninvasive visualization of tumor lesions.
Collapse
Affiliation(s)
- Lixin Ding
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Research, Investigation and Evaluation of Radiopharmaceuticals, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Qian Guo
- Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, China
| | - Ya'nan Ren
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Research, Investigation and Evaluation of Radiopharmaceuticals, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Pei Wang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Research, Investigation and Evaluation of Radiopharmaceuticals, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Yongxiang Pan
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Research, Investigation and Evaluation of Radiopharmaceuticals, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Jin Ding
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Research, Investigation and Evaluation of Radiopharmaceuticals, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Feng Wang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Research, Investigation and Evaluation of Radiopharmaceuticals, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Zhi Yang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Research, Investigation and Evaluation of Radiopharmaceuticals, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Hua Zhu
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Research, Investigation and Evaluation of Radiopharmaceuticals, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing 100142, China
| |
Collapse
|
5
|
Su Y, He W, Zheng L, Fan X, Hu TY. Toward Clarity in Single Extracellular Vesicle Research: Defining the Field and Correcting Missteps. ACS NANO 2025; 19:16193-16203. [PMID: 40271998 PMCID: PMC12060644 DOI: 10.1021/acsnano.5c00705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/12/2025] [Revised: 04/11/2025] [Accepted: 04/11/2025] [Indexed: 04/25/2025]
Abstract
Single extracellular vesicle (EV) research holds the potential to revolutionize our understanding of cellular communication and enable breakthroughs in diagnostics and therapeutics. However, the lack of a clear, consensus-driven definition of single EV research has led to methodological inconsistencies, overgeneralized interpretations, and, in some cases, misleading claims. In this perspective, we propose a framework for defining single EV research, critique current challenges and misconceptions in this field, and discuss its implications for biomedical applications. We argue that precise experimental design, rigorous validation, and interdisciplinary collaboration approaches are needed to establish single EV research as a cornerstone of precision medicine.
Collapse
Affiliation(s)
- Yun Su
- Department
of Ophthalmology, Shanghai Key Laboratory of Orbital Diseases and
Ocular Oncology, Shanghai Ninth People’s
Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, P. R. China
| | - Wanzhuo He
- Department
of Ophthalmology, Shanghai Key Laboratory of Orbital Diseases and
Ocular Oncology, Shanghai Ninth People’s
Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, P. R. China
| | - Lei Zheng
- Department
of Laboratory Medicine, Guangdong Provincial Key Laboratory of Precision
Medical Diagnostics, Guangdong Engineering and Technology Research
Center for Rapid Diagnostic Biosensors, Guangdong Provincial Key Laboratory
of Single Cell Technology and Application, Nanfang Hospital, Southern Medical University, Guangzhou 510515, P. R. China
| | - Xianqun Fan
- Department
of Ophthalmology, Shanghai Key Laboratory of Orbital Diseases and
Ocular Oncology, Shanghai Ninth People’s
Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, P. R. China
| | - Tony Y. Hu
- Department
of Biochemistry and Molecular Biology, Center for Cellular and Molecular
Diagnostics, Tulane University School of
Medicine, New Orleans, Louisiana 70112, United States
| |
Collapse
|
6
|
Aksamitiene E, Park J, Marjanovic M, Boppart SA. Defining Biological Variability, Analytical Precision and Quantitative Biophysiochemical Characterization of Human Urinary Extracellular Vesicles. J Extracell Vesicles 2025; 14:e70087. [PMID: 40384173 PMCID: PMC12086329 DOI: 10.1002/jev2.70087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Accepted: 04/18/2025] [Indexed: 05/20/2025] Open
Abstract
The magnitude of combined analytical errors of urinary extracellular vesicle (uEV) preparation and measurement techniques (CVA) has not been thoroughly investigated to determine whether it exceeds biological variations. We utilized technical replicates of human urine to assess the repeatability of uEV concentration and size measurements by nanoparticle tracking analysis (NTA) following differential velocity centrifugation (DC), silicon carbide, or polyethylene glycol uEV isolation methods. The DC method attained the highest precision. Consequently, DC-derived uEV size, most abundant protein levels, and optical redox ratio (ORR) were further assessed by dynamic light scattering (DLS), immunoblotting or multi-photon (SLAM) microscopy. Procedural errors primarily affected uEV counting and uEV-associated protein quantification, while instrumental errors contributed most to the total variability of NTA- and DLS-mediated uEV sizing processes. The intra-individual variability (CVI) of uEV counts assessed by NTA was smaller than inter-individual variability (CVG), resulting in an estimated index of individuality IOI < 0.6, suggesting that personalized reference interval (RI) is more suitable for interpretation of changes in subject's test results. Population-based RI was more appropriate for ORR (IOI > 1.4). The analytical performance of DC-NTA and DC-SLAM techniques met optimal CVA < 0.5 × CVI criteria, indicating their suitability for further testing in clinical laboratory settings.
Collapse
Affiliation(s)
- Edita Aksamitiene
- Beckman Institute for Advanced Science and TechnologyUniversity of Illinois Urbana‐ChampaignUrbanaIllinoisUSA
- NIH/NIBIB P41 Center for Label‐Free Imaging and Multiscale BiophotonicsUniversity of Illinois Urbana‐ChampaignUrbanaIllinoisUSA
| | - Jaena Park
- Beckman Institute for Advanced Science and TechnologyUniversity of Illinois Urbana‐ChampaignUrbanaIllinoisUSA
- NIH/NIBIB P41 Center for Label‐Free Imaging and Multiscale BiophotonicsUniversity of Illinois Urbana‐ChampaignUrbanaIllinoisUSA
- Department of BioengineeringUniversity of Illinois Urbana‐ChampaignUrbanaIllinoisUSA
| | - Marina Marjanovic
- Beckman Institute for Advanced Science and TechnologyUniversity of Illinois Urbana‐ChampaignUrbanaIllinoisUSA
- NIH/NIBIB P41 Center for Label‐Free Imaging and Multiscale BiophotonicsUniversity of Illinois Urbana‐ChampaignUrbanaIllinoisUSA
| | - Stephen A. Boppart
- Beckman Institute for Advanced Science and TechnologyUniversity of Illinois Urbana‐ChampaignUrbanaIllinoisUSA
- NIH/NIBIB P41 Center for Label‐Free Imaging and Multiscale BiophotonicsUniversity of Illinois Urbana‐ChampaignUrbanaIllinoisUSA
- Department of BioengineeringUniversity of Illinois Urbana‐ChampaignUrbanaIllinoisUSA
- Department of Electrical and Computer EngineeringUniversity of Illinois Urbana‐ChampaignUrbanaIllinoisUSA
- Cancer Center at IllinoisUniversity of Illinois Urbana‐ChampaignUrbanaIllinoisUSA
- Carle Illinois College of MedicineUniversity of Illinois Urbana‐ChampaignUrbanaIllinoisUSA
- Interdisciplinary Health Sciences InstituteUniversity of Illinois Urbana‐ChampaignUrbanaIllinoisUSA
| |
Collapse
|
7
|
Rajendran RL, Gangadaran P, Ghosh S, Nagarajan AK, Batabyal R, Ahn BC. Unlocking the secrets of single extracellular vesicles by cutting-edge technologies. Pathol Res Pract 2025; 269:155878. [PMID: 40024075 DOI: 10.1016/j.prp.2025.155878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 02/20/2025] [Accepted: 02/27/2025] [Indexed: 03/04/2025]
Abstract
Extracellular vesicles (EVs), isolated through techniques such as liquid biopsy, have emerged as crucial biomarkers in various diseases, including cancer. EVs were dismissed initially as cellular debris, EVs are now recognized for their role in intercellular communication, carrying proteins, RNAs, and other molecules between cells. Their stability in biofluids and ability to mirror their parent cells' molecular composition make them attractive candidates for non-invasive diagnostics. EVs, including microvesicles and exosomes, contribute to immune modulation and cancer progression, presenting both therapeutic challenges and opportunities. However, despite advances in analytical techniques like high-resolution microscopy and nanoparticle tracking analysis (NTA), standardization in EV isolation and characterization remains a hurdle. Cutting-edge technologies, such as atomic force microscopy and Raman tweezers microspectroscopy, have enhanced our understanding of single EVs, yet issues like low throughput and high technical complexity limit their widespread application. Other technologies like transmission electron microscopy, cryogenic transmission electron microscopy, super-resolution microscopy, direct stochastic optical reconstruction microscopy, single-molecule localization microscopy, tunable resistive pulse sensing, single-particle interferometric reflectance imaging sensor, flow cytometry, droplet digital analysis, total internal reflection fluorescence also contribute to EV analysis. Future research must focus on improving detection methods, developing novel analytical platforms, and integrating artificial intelligence to enhance the specificity of EV characterization. The future of EV research holds promise for breakthroughs in precision medicine, with a collaborative effort needed to translate these advancements into clinical practice.
Collapse
Affiliation(s)
- Ramya Lakshmi Rajendran
- BK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Department of Biomedical Science, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea; Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea; Cardiovascular Research Institute, Kyungpook National University, Daegu 41944, Republic of Korea
| | - Prakash Gangadaran
- BK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Department of Biomedical Science, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea; Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea; Cardiovascular Research Institute, Kyungpook National University, Daegu 41944, Republic of Korea
| | - Subhrojyoti Ghosh
- Department of Biotechnology, Indian Institute of Technology, Madras, Chennai 600036, India
| | - ArulJothi Kandasamy Nagarajan
- Department of Genetic Engineering, SRM Institute of Science and Technology, Kattankulathur, Chengalpattu, Tamilnadu 603203, India
| | - Rijula Batabyal
- Department of Biotechnology, Heritage Institute of Technology, Kolkata 700 107, India
| | - Byeong-Cheol Ahn
- BK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Department of Biomedical Science, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea; Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea; Cardiovascular Research Institute, Kyungpook National University, Daegu 41944, Republic of Korea; Department of Nuclear Medicine, Kyungpook National University Hospital, Daegu 41944, Republic of Korea.
| |
Collapse
|
8
|
Glamočlija S, Sabljić L, Tomić S, Đokić J, Radulović N, Gruden-Movsesijan A, Kosanović M. Trichinella spiralis extracellular vesicles induce anti-inflammatory and regulatory immune responses in vitro. Int J Parasitol 2025; 55:299-315. [PMID: 39842685 DOI: 10.1016/j.ijpara.2025.01.008] [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: 07/30/2024] [Revised: 10/21/2024] [Accepted: 01/14/2025] [Indexed: 01/24/2025]
Abstract
The helminth Trichinella spiralis, through its excretory-secretory (ES L1) products, induces immune regulatory mechanisms that modulate the host's immune response not only to itself, but also to bystander antigens, foreign or self in origin, which can result in the alleviation of inflammatory diseases. Under the influence of ES L1, dendritic cells (DCs) acquire a tolerogenic phenotype and the capacity to induce Th2 and regulatory responses. Since ES L1 products represent a complex mixture of proteins and extracellular vesicles (TsEVs) the aim of this study was to investigate the impact of TsEVs, isolated from ES L1 products, on phenotypic and functional characteristics of DCs and to elucidate whether TsEVs could reproduce the immunomodulatory effects of the complete ES L1 product. Monocyte-derived DCs treated with TsEVs acquired semi-matured phenotypes, characterized by low expression of human leukocyte antigen - DR isotype (HLA-DR), cluster of differentiation (CD) 86 (CD86), and CD40, moderate expression of CD83 and C-C chemokine receptor type 7 (CCR7), and increased expression of tolerogenic markers indoleamine 2,3-dioxygenase 1 (IDO-1) and immunoglobulin-like transcript 3 (ILT3), together with the unchanged production of IL-12 and IL-23, and elevated production of IL-10 and transforming growth factor (TGF)-β, compared with controls. Gene expression analysis of TsEV-treated DCs revealed elevated levels of mTOR, Ahr, NF-κB2, RelB, SOCS1 and SOCS3, which participate in signaling pathways involved in DC maturation and the subsequent regulation of release of both anti-inflammatory and pro-inflammatory cytokines. TsEVs promoted the capacity of DCs to drive polarization of Th2 and anti-inflammatory responses, and impaired their capacity to induce Th1/Th17 polarization. Moreover, TsEV-treated DCs possessed a high capacity to induce conventional FoxP3 + regulatory T cells, as well as unconventional T regulatory (Tr1) cells. Tolerogenic properties of TsEV-treated DCs were retained even after challenge with a pro-inflammatory stimulus. These findings highlight the potential of TsEVs to induce immune tolerance, suggesting their potential use as therapeutics for the treatment of inflammatory disorders.
Collapse
Affiliation(s)
- Sofija Glamočlija
- Institute for the Application of Nuclear Energy INEP University of Belgrade Republic of Serbia
| | - Ljiljana Sabljić
- Institute for the Application of Nuclear Energy INEP University of Belgrade Republic of Serbia
| | - Sergej Tomić
- Institute for the Application of Nuclear Energy INEP University of Belgrade Republic of Serbia
| | - Jelena Đokić
- Institute of Molecular Genetics and Genetic Engineering IMGGE University of Belgrade Republic of Serbia
| | - Nataša Radulović
- Institute for Biological Research "Siniša Stanković" University of Belgrade Republic of Serbia
| | - Alisa Gruden-Movsesijan
- Institute for the Application of Nuclear Energy INEP University of Belgrade Republic of Serbia
| | - Maja Kosanović
- Institute for the Application of Nuclear Energy INEP University of Belgrade Republic of Serbia.
| |
Collapse
|
9
|
Escudero-Cernuda S, Eiro N, Fraile M, Vizoso FJ, Fernández-Colomer B, Fernández-Sánchez ML. Limitations and challenges in the characterization of extracellular vesicles from stem cells and serum. Mikrochim Acta 2025; 192:311. [PMID: 40259021 PMCID: PMC12011935 DOI: 10.1007/s00604-025-07147-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Accepted: 04/01/2025] [Indexed: 04/23/2025]
Abstract
Exosomes are a subpopulation of nanosized extracellular vesicles (EVs), formed by a lipid bilayer and naturally secreted by cells. They transport RNA, microRNAs, bioactive proteins, and lipids and play an important role in intercellular communication. Exosomes are a promising alternative cell-free therapy in regenerative medicine, immunotherapy, and drug delivery. The implementation of new exosomes treatments requires knowledge of its concentration, purity, and full characterization. However, comparing different studies is highly challenging due to the lack of validated methodologies for isolation and determination, as well as a lack of well-characterized exosomes reference standards. In this work, human uterine cervical mesenchymal stem cell (hUCESC) EVs have been isolated by ultracentrifugation and characterized, discussing the current limitations of characterization methods. First, total protein assays are heavily influenced by free-protein and lipid contaminations which confirm the need of employing several methods for vesicular protein determination. This purity variation seems heavily influenced by the vesicles origin as more complex mediums originate more matrix interferences. Size exclusion high performance liquid chromatography has been demonstrated as a new methodology for purity assessment of hUCESC-EVs and commercial EVs (adipose stem cells and human serum). The results found low purity in the commercial exosomes highlighting that protein and lipid purity must be included in the commercial EVs. Finally, the combination of this chromatography method with total protein assays proved that particle concentration could be estimated using vesicular protein concentration.
Collapse
Affiliation(s)
- Sara Escudero-Cernuda
- Department of Physical and Analytical Chemistry, University of Oviedo, Avda. Julián Clavería, 8, 33006, Oviedo, Asturias, Spain
| | - Noemi Eiro
- Research Unit, Jove Hospital Foundation, Avda. Eduardo Castro, 161, 33920, Gijón, Asturias, Spain.
| | - María Fraile
- Research Unit, Jove Hospital Foundation, Avda. Eduardo Castro, 161, 33920, Gijón, Asturias, Spain
| | - Francisco J Vizoso
- Research Unit, Jove Hospital Foundation, Avda. Eduardo Castro, 161, 33920, Gijón, Asturias, Spain.
| | - Belén Fernández-Colomer
- Service of Neonatology, Department of Pediatrics, Hospital Universitario Central de Asturias, Oviedo, Spain
| | - María Luisa Fernández-Sánchez
- Department of Physical and Analytical Chemistry, University of Oviedo, Avda. Julián Clavería, 8, 33006, Oviedo, Asturias, Spain.
| |
Collapse
|
10
|
Ozerklig B, Turkel I, Yilmaz M, Vaizoglu RD, Akan HS, Dikmen ZG, Saleem A, Kosar SN. Exercise-induced extracellular vesicles mediate apoptosis in human colon cancer cells in an exercise intensity-dependent manner. Eur J Appl Physiol 2025:10.1007/s00421-025-05787-1. [PMID: 40253655 DOI: 10.1007/s00421-025-05787-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Accepted: 03/26/2025] [Indexed: 04/22/2025]
Abstract
Regular exercise reduces the incidence and improves the prognosis of many cancer types, but the underlying mechanisms remain elusive. Evidence suggests that exercise exerts its therapeutic effects through extracellular vesicles (EVs), which are essential for cellular communication. Here, we hypothesized that exercise-induced EVs from serum of healthy individuals would exert anti-tumorigenic effects on human colon cancer HT-29 cells, in an exercise intensity-dependent manner. Ten healthy young active males participated in a randomized crossover trial, completing two workload-matched acute exercise bouts, moderate-intensity continuous exercise (MICE) and high-intensity interval exercise (HIIE), on a cycle ergometer. A control session of rest (PRE) was included. EVs were isolated from serum samples collected during PRE and immediately after each exercise session. EVs were co-incubated with HT-29 colon cancer cells, and the effects on cell viability, migration, and apoptosis were measured. EV treatment reduced cell viability in all groups (PRE, MICE, and HIIE) by 35%, 43% and 47%, respectively, vs. PBS. HIIE-EVs showed a significantly greater reduction in cell viability vs. PRE; therefore, only these groups were used for further analysis. PRE EVs reduced migration by 27%, and HIIE-EVs by 39%. HIIE-EVs increased expression of pro-apoptotic markers: Bax/Bcl-2 ratio by 56% and Caspase 3 by 30% vs. PBS, with no change observed in the PRE group. Further, 16% of cells in PRE and 28% of cells in HIIE were TUNEL-positive, indicating DNA fragmentation. To our knowledge, this is the first human study that illustrates the therapeutic potential of exercise-induced EVs in cancer treatment.
Collapse
Affiliation(s)
- Berkay Ozerklig
- Department of Exercise and Sport Sciences, Faculty of Sport Sciences, Hacettepe University, Ankara, Türkiye.
- Faculty of Kinesiology and Recreation Management, University of Manitoba, Winnipeg, Canada.
- The Children's Hospital Research Institute of Manitoba (CHRIM), Winnipeg, Canada.
| | - Ibrahim Turkel
- Department of Exercise and Sport Sciences, Faculty of Sport Sciences, Hacettepe University, Ankara, Türkiye
| | - Merve Yilmaz
- Department of Medical Biochemistry, Faculty of Medicine, Hacettepe University, Ankara, Türkiye
| | - Refika Dilara Vaizoglu
- Department of Biology, Molecular Biology Section, Faculty of Science, Hacettepe University, Ankara, Türkiye
| | - Handan Sevim Akan
- Department of Biology, Faculty of Science, Hacettepe University, Ankara, Türkiye
| | - Z Gunnur Dikmen
- Department of Medical Biochemistry, Faculty of Medicine, Hacettepe University, Ankara, Türkiye
| | - Ayesha Saleem
- Faculty of Kinesiology and Recreation Management, University of Manitoba, Winnipeg, Canada
- The Children's Hospital Research Institute of Manitoba (CHRIM), Winnipeg, Canada
| | - Sukran Nazan Kosar
- Division of Exercise Nutrition and Metabolism, Faculty of Sport Sciences, Hacettepe University, Ankara, Türkiye
| |
Collapse
|
11
|
Braunsperger MV, Martin G, Herzig T, Kußberger I, Gießl A, Steimle S, Schlötzer-Schrehardt U, Schlunck G, Reinhard T, Polisetti N. Proteomic Insights into Human Limbal Epithelial Progenitor-Derived Small Extracellular Vesicles. Stem Cell Rev Rep 2025:10.1007/s12015-025-10877-w. [PMID: 40238075 DOI: 10.1007/s12015-025-10877-w] [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: 04/04/2025] [Indexed: 04/18/2025]
Abstract
Limbal epithelial stem/progenitor cells (LEPC), supported by limbal mesenchymal stromal cells (LMSC) and limbal melanocytes (LM) within a specialized niche, are responsible for maintaining the corneal epithelium. Small extracellular vesicles (sEV) emerged as critical mediators of intercellular communication in various stem cell niches, yet their role in maintaining human limbal niche homeostasis remains poorly understood. In this study, tangential flow filtration and size exclusion chromatography were used to isolate sEV from LEPC-, LMSC- and LM-conditioned media. The isolated sEV from LEPC exhibited properties characteristic for sEV as confirmed by nanoparticle tracking analysis for size and concentration, by electron microscopy for morphology, and by western blot analysis of canonical EV markers including the cell-specific protein (cytokeratin 17/19). Quantitative and comparative proteomic profiling revealed distinct molecular signatures of LEPC-derived sEV, enriched in factors associated with keratinocyte development, extracellular matrix organization, and niche regulation. These findings suggest that LEPC-derived sEV may serve as important signaling mediators within the limbal niche microenvironment, though additional studies are needed to determine their specific functional roles in maintaining niche homeostasis.
Collapse
Affiliation(s)
- Moritz Vincent Braunsperger
- Eye Center, Medical Center - Faculty of Medicine, University of Freiburg, Killianstrasse 5, 79106, Freiburg, Germany
| | - Gottfried Martin
- Eye Center, Medical Center - Faculty of Medicine, University of Freiburg, Killianstrasse 5, 79106, Freiburg, Germany
| | - Tabea Herzig
- Eye Center, Medical Center - Faculty of Medicine, University of Freiburg, Killianstrasse 5, 79106, Freiburg, Germany
| | - Isabell Kußberger
- Eye Center, Medical Center - Faculty of Medicine, University of Freiburg, Killianstrasse 5, 79106, Freiburg, Germany
| | - Andreas Gießl
- Department of Ophthalmology, University Hospital Erlangen, Friedrich-Alexander-University of Erlan-gen-Nürnberg, Schwabachanlage 6, D-91054, Erlangen, Germany
| | - Stefan Steimle
- Cryo-EM Facility (CEF), University of Freiburg, Albertstrasse 21, 79104, Freiburg, Germany
- Institute of Physical Chemistry, University of Freiburg, Albertstrasse 21, 79104, Freiburg, Germany
| | - Ursula Schlötzer-Schrehardt
- Department of Ophthalmology, University Hospital Erlangen, Friedrich-Alexander-University of Erlan-gen-Nürnberg, Schwabachanlage 6, D-91054, Erlangen, Germany
| | - Günther Schlunck
- Eye Center, Medical Center - Faculty of Medicine, University of Freiburg, Killianstrasse 5, 79106, Freiburg, Germany
| | - Thomas Reinhard
- Eye Center, Medical Center - Faculty of Medicine, University of Freiburg, Killianstrasse 5, 79106, Freiburg, Germany
| | - Naresh Polisetti
- Eye Center, Medical Center - Faculty of Medicine, University of Freiburg, Killianstrasse 5, 79106, Freiburg, Germany.
| |
Collapse
|
12
|
Isik M, Sari HK, Caglayan MG, Yilmaz R, Derkus B. Whispers in the Brain: Extracellular Vesicles in Neuropathology and the Diagnostic Alchemy of Neurological Diseases. Eur J Neurosci 2025; 61:e70090. [PMID: 40237381 DOI: 10.1111/ejn.70090] [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: 09/23/2024] [Revised: 02/21/2025] [Accepted: 03/15/2025] [Indexed: 04/18/2025]
Abstract
Extracellular vesicles (EVs) have emerged as pivotal mediators in neurological diseases, showcasing multifaceted potential roles ranging from pathogenesis to diagnosis. These nano-sized membranous structures, released by various cell types including neurons, astrocytes, and microglia, encapsulate a diverse cargo of proteins, lipids, RNA species, and even DNA fragments. In neuropathology, EVs contribute significantly to intercellular communication within the central nervous system (CNS), influencing physiological or pathological cascades. Through the transfer of bioactive molecules, EVs modulate neuroinflammation, neuronal survival, synaptic plasticity, and the propagation of protein aggregates characteristic of neurodegenerative disorders. Moreover, their presence in biofluids such as cerebrospinal fluid (CSF), blood, and urine reflects the pathophysiological state of the CNS, offering a window into the diagnosis, monitoring and treatment of neurological diseases. Recent advancements in EV isolation techniques, coupled with high-throughput omics technologies, have facilitated the profiling of EV cargo, enabling the identification of disease-specific biomarkers with high sensitivity and specificity. This review explores the intricate roles of EVs in neuropathology, highlighting their involvement in Alzheimer's disease, Parkinson's disease, multiple sclerosis, and other neurological disorders. Furthermore, it delves into the diagnostic potential of EVs, discussing current challenges and prospects in harnessing EV-derived biomarkers for precision medicine in neurology. Ultimately, understanding the biology of EVs in neurological contexts promises transformative insights into disease mechanisms and therapeutic strategies, paving the way for innovative diagnostic tools and targeted interventions in clinical practice.
Collapse
Affiliation(s)
- Melis Isik
- Stem Cell Research Lab, Department of Chemistry, Faculty of Science, Ankara University, Ankara, Turkey
| | - Hatice Kubra Sari
- Department of Neurology, School of Medicine, Ankara University, Ankara, Turkey
| | - Mehmet Gokhan Caglayan
- Department of Analytical Chemistry, Faculty of Pharmacy, Ankara University, Ankara, Turkey
| | - Rezzak Yilmaz
- Department of Neurology, School of Medicine, Ankara University, Ankara, Turkey
- Brain Research Center, Ankara University, Ankara, Turkey
| | - Burak Derkus
- Stem Cell Research Lab, Department of Chemistry, Faculty of Science, Ankara University, Ankara, Turkey
- Neuroscience and Neurotechnology Excellence Joint Application and Research Center (NEUROM), Ankara, Turkey
| |
Collapse
|
13
|
Wang WB, Wan JY, Yu DJ, Du HX, Zhou HF, Wan HT, Yang JH. Chlorogenic acid inhibits virulence and resistance gene transfer in outer membrane vesicles of carbapenem-resistant Klebsiella pneumoniae. Front Pharmacol 2025; 16:1562096. [PMID: 40230687 PMCID: PMC11994928 DOI: 10.3389/fphar.2025.1562096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Accepted: 03/07/2025] [Indexed: 04/16/2025] Open
Abstract
Introduction Carbapenem-resistant Klebsiella pneumoniae (CRKp) infection poses a significant global public health challenge, with the misuse of antibiotics further contributing to the development of resistance and triggering harmful inflammatory responses. Outer membrane vesicles (OMVs) released by CRKp under sub-lethal concentration of MEM pressure (KOMV-MEM) exhibit enhanced virulence and greater efficiency in transferring resistance genes. Methods We investigated the inhibitory effects of chlorogenic acid (CA) on KOMV-MEM characteristics and its protective role in KOMV-MEM infected mice. Based on LC-MS proteomic analysis of vesicles, we screened for potential targets of KOMV-MEM in promoting macrophage (MØ) pyroptosis pathways and inducing resistance gene transfer. Subsequently, computational predictions and experimental validation were performed to determine how CA regulates these mechanisms. Results This study confirmed that, under MEM pressure, the exacerbated infection levels in CRKp-inoculated mice are attributable to the high virulence of KOMV-MEM. Computational and experimental results demonstrated that CA inhibits pyroptosis by reducing MØ capture of KOMV-MEM through blocking the interaction between GroEL and LOX-1. Furthermore, CA prevents the spread of resistance genes by disrupting the conjugation and transfer processes between KOMV-MEM and recipient bacteria. Finally, in vitro and in vivo assays showed that CA inhibits KOMV-MEM resistance enzymes, thereby preventing the hydrolysis of MEM in the environment and depriving susceptible bacteria of protection. Discussion These findings provide the first confirmation that CA can inhibit both the virulence and the transmission of drug resistance in KOMV-MEM. This underscores the potential of CA treatment as a promising antimicrobial strategy against CRKp infection.
Collapse
Affiliation(s)
- Wen-Ba Wang
- College of Basic Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jia-Yang Wan
- The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Dao-Jun Yu
- Department of Medical Laboratory, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Hai-Xia Du
- College of Basic Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Hui-Fen Zhou
- College of Basic Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Hai-Tong Wan
- College of Basic Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jie-Hong Yang
- College of Basic Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
14
|
Farm YR, Chuah BH, Law JX, Leong XF, Razali M, Ng SL. Therapeutic Potential of Extracellular Vesicles in Oral Inflammation. Int J Mol Sci 2025; 26:3031. [PMID: 40243684 PMCID: PMC11988662 DOI: 10.3390/ijms26073031] [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: 02/21/2025] [Revised: 03/18/2025] [Accepted: 03/22/2025] [Indexed: 04/18/2025] Open
Abstract
The therapeutic potential of extracellular vesicles (EVs) in reducing oral inflammation is thoroughly examined in this review, with an emphasis on gingivitis, periodontitis, and oral mucositis. It explains the complex relationship between microbial dysbiosis and host immune responses in the aetiology of oral inflammation. Pathophysiological mechanisms of periodontitis are examined, emphasising the roles played by periodontal pathogens and inflammatory mediators in the disease's chronic course and systemic effects. Preclinical research is providing new evidence that EVs originating from various cellular sources control immune cell dynamics towards a pro-healing phenotype, promote tissue regeneration, and have immunomodulatory qualities. EV-based therapies appear to be a promising new therapeutic technique with potential benefits over traditional methods for the treatment of oral inflammatory illnesses by specifically altering inflammatory signalling pathways. This review highlights the potential of EVs to improve patient outcomes in oral health and emphasises the need for additional clinical research to clarify the therapeutic efficacy and underlying mechanisms of EVs in periodontal therapy.
Collapse
Affiliation(s)
- Yan Rou Farm
- Department of Craniofacial Diagnostics and Biosciences, Faculty of Dentistry, Universiti Kebangsaan Malaysia, Kuala Lumpur 50300, Malaysia; (Y.R.F.); (B.H.C.); (X.F.L.)
| | - Bing Huan Chuah
- Department of Craniofacial Diagnostics and Biosciences, Faculty of Dentistry, Universiti Kebangsaan Malaysia, Kuala Lumpur 50300, Malaysia; (Y.R.F.); (B.H.C.); (X.F.L.)
| | - Jia Xian Law
- Department of Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia;
| | - Xin Fang Leong
- Department of Craniofacial Diagnostics and Biosciences, Faculty of Dentistry, Universiti Kebangsaan Malaysia, Kuala Lumpur 50300, Malaysia; (Y.R.F.); (B.H.C.); (X.F.L.)
| | - Masfueh Razali
- Department of Restorative Dentistry, Faculty of Dentistry, Universiti Kebangsaan Malaysia, Kuala Lumpur 50300, Malaysia;
| | - Sook Luan Ng
- Department of Craniofacial Diagnostics and Biosciences, Faculty of Dentistry, Universiti Kebangsaan Malaysia, Kuala Lumpur 50300, Malaysia; (Y.R.F.); (B.H.C.); (X.F.L.)
| |
Collapse
|
15
|
Cirillo C, Iuliano M, Fierro F, Florio C, Maffei G, Loi A, Batakliev T, Sarno M. Silver Nanoparticle-Based Finishing for Leather Antimicrobial and UV Protection. MICROMACHINES 2025; 16:376. [PMID: 40283253 PMCID: PMC12029507 DOI: 10.3390/mi16040376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/26/2025] [Revised: 03/21/2025] [Accepted: 03/23/2025] [Indexed: 04/29/2025]
Abstract
This study focuses on preparing and characterizing functionalized silver nanoparticle-based (Ag-F NPs) finishing agents for leather treatment. Ag-F NPs were synthesized and functionalized through a ligand exchange process with citric acid, enhancing their dispersion stability in aqueous media. The nanoparticles were incorporated into polyurethane- and nitroemulsion-based finishing formulations and applied to ovine and bovine leather via a spray coating process. Morphological (SEM, TEM), structural (XRD), thermal (TGA), and spectroscopic (FT-IR) analyses confirmed successful functionalization and uniform dispersion within the finishing layer. Leather samples treated with Ag-F NPs exhibited a significant improvement in antibacterial properties, with microbial growth reduction of up to 90% after 72 h. Additionally, accelerated aging tests demonstrated enhanced UV resistance, with a 30% lower color change (∆E) compared to control samples. The Ag-F NPs-based finishing layers also exhibited superior abrasion and micro-scratch resistance, maintaining a stable coefficient of friction over time. These findings demonstrate the potential of Ag-F NPs as multifunctional leather-finishing agents, making them highly suitable for applications in the automotive, footwear, and leather goods industries.
Collapse
Affiliation(s)
- Claudia Cirillo
- Department of Physics “E.R. Caianiello”, University of Salerno, Via Giovanni Paolo II, 132, 84084 Fisciano, Italy; (M.I.)
- Centre NANO_MATES, University of Salerno, Via Giovanni Paolo II, 132, 84084 Fisciano, Italy
| | - Mariagrazia Iuliano
- Department of Physics “E.R. Caianiello”, University of Salerno, Via Giovanni Paolo II, 132, 84084 Fisciano, Italy; (M.I.)
- Centre NANO_MATES, University of Salerno, Via Giovanni Paolo II, 132, 84084 Fisciano, Italy
| | - Francesca Fierro
- Department of Physics “E.R. Caianiello”, University of Salerno, Via Giovanni Paolo II, 132, 84084 Fisciano, Italy; (M.I.)
- Centre NANO_MATES, University of Salerno, Via Giovanni Paolo II, 132, 84084 Fisciano, Italy
| | - Claudia Florio
- Stazione Sperimentale per l’Industria delle Pelli e delle Materie Concianti—SSIP (Italian Leather Research Institute), Comprensorio Olivetti, Via Campi Flegrei, 34, 80078 Pozzuoli, Italy
| | - Gaetano Maffei
- Conceria DMD SOLOFRA S.p.A., Via Celentane, 9, 83029 Solofra, Italy
| | - Andrea Loi
- Mario Levi Italia S.r.l., Via Arzignano, 130, 36072 Chiampo, Italy
| | - Todor Batakliev
- Open Laboratory on Experimental Micro and Nano Mechanics (OLEM), Institute of Mechanics, Bulgarian Academy of Sciences, Acad. G. Bonchev Str., Block 4, 1113 Sofia, Bulgaria
| | - Maria Sarno
- Department of Physics “E.R. Caianiello”, University of Salerno, Via Giovanni Paolo II, 132, 84084 Fisciano, Italy; (M.I.)
- Centre NANO_MATES, University of Salerno, Via Giovanni Paolo II, 132, 84084 Fisciano, Italy
| |
Collapse
|
16
|
Schorr K, Beck S, Zimmer O, Baumann F, Keller M, Witzgall R, Goepferich A. The quantity of ligand-receptor interactions between nanoparticles and target cells. NANOSCALE HORIZONS 2025; 10:803-823. [PMID: 39951050 DOI: 10.1039/d4nh00645c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/25/2025]
Abstract
Achieving high target cell avidity in combination with cell selectivity are fundamental, but largely unachieved goals in the development of biomedical nanoparticle systems, which are intricately linked to the quantity of targeting functionalities on their surface. Viruses, regarded as almost ideal role models for nanoparticle design, are evolutionary optimized, so that they cope with this challenge bearing an extremely low number of spikes, and thus binding domains, on their surface. In comparison, nanoparticles are usually equipped with more than an order of magnitude more ligands. It is therefore obvious that one key factor for increasing nanoparticle efficiency in terms of avidity and selectivity lies in optimizing their ligand number. A first step along this way is to know how many ligands per nanoparticle are involved in specific binding with target cell receptors. This question is addressed experimentally for a block copolymer nanoparticle model system. The data confirm that only a fraction of the nanoparticle ligands is involved in the binding processes: with a total ligand valency of 29 ligands/100 nm2 surface area a maximum 5.3 ligands/100 nm2 are involved in specific receptor binding. This corresponds to an average number of 251 binding ligands per nanoparticle, a number that can be rationalized within the biological context of the model system.
Collapse
Affiliation(s)
- Kathrin Schorr
- Department of Pharmaceutical Technology, University of Regensburg, 93053 Regensburg, Bavaria, Germany.
| | - Sebastian Beck
- Department of Pharmaceutical Technology, University of Regensburg, 93053 Regensburg, Bavaria, Germany.
| | - Oliver Zimmer
- Department of Pharmaceutical Technology, University of Regensburg, 93053 Regensburg, Bavaria, Germany.
| | - Felix Baumann
- Department of Pharmaceutical Technology, University of Regensburg, 93053 Regensburg, Bavaria, Germany.
| | - Max Keller
- Institute of Pharmacy, Faculty of Chemistry and Pharmacy, University of Regensburg, D-93040 Regensburg, Germany.
| | - Ralph Witzgall
- Institute for Molecular and Cellular Anatomy, University of Regensburg, 93053 Regensburg, Bavaria, Germany.
| | - Achim Goepferich
- Department of Pharmaceutical Technology, University of Regensburg, 93053 Regensburg, Bavaria, Germany.
| |
Collapse
|
17
|
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
|
18
|
Puagsopa J, Tongviseskul N, Jaroentomeechai T, Meksiriporn B. Recent Progress in Developing Extracellular Vesicles as Nanovehicles to Deliver Carbohydrate-Based Therapeutics and Vaccines. Vaccines (Basel) 2025; 13:285. [PMID: 40266147 PMCID: PMC11946770 DOI: 10.3390/vaccines13030285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 02/22/2025] [Accepted: 03/04/2025] [Indexed: 04/24/2025] Open
Abstract
Cell-derived, nanoscale extracellular vesicles (EVs) have emerged as promising tools in diagnostic, therapeutic, and vaccine applications. Their unique properties including the capability to encapsulate diverse molecular cargo as well as the versatility in surface functionalization make them ideal candidates for safe and effective vehicles to deliver a range of biomolecules including gene editing cassettes, therapeutic proteins, glycans, and glycoconjugate vaccines. In this review, we discuss recent advances in the development of EVs derived from mammalian and bacterial cells for use in a delivery of carbohydrate-based protein therapeutics and vaccines. We highlight key innovations in EVs' molecular design, characterization, and deployment for treating diseases including Alzheimer's disease, infectious diseases, and cancers. We discuss challenges for their clinical translation and provide perspectives for future development of EVs within biopharmaceutical research and the clinical translation landscape.
Collapse
Affiliation(s)
- Japigorn Puagsopa
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, FL 32610, USA;
| | - Niksa Tongviseskul
- Department of Biology, School of Science, King Mongkut’s Institute of Technology Ladkrabang, Bangkok 10520, Thailand;
| | - Thapakorn Jaroentomeechai
- Copenhagen Center for Glycomics, Departments of Cellular and Molecular Medicine, Faculty of Health Sciences, University of Copenhagen, Blegdamsvej 3, 2200 Copenhagen, Denmark
| | - Bunyarit Meksiriporn
- Department of Biology, School of Science, King Mongkut’s Institute of Technology Ladkrabang, Bangkok 10520, Thailand;
| |
Collapse
|
19
|
Feix AS, Laimer-Digruber A, Cruz-Bustos T, Steiner G, Ruttkowski B, Ehling-Schulz M, Joachim A. Variations in extracellular vesicle shedding of Cystoisospora suis stages (Apicomplexa: Coccidia). Int J Parasitol 2025; 55:197-212. [PMID: 39793881 DOI: 10.1016/j.ijpara.2025.01.001] [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: 06/12/2024] [Revised: 11/21/2024] [Accepted: 01/05/2025] [Indexed: 01/13/2025]
Abstract
Cystoisospora suis, a porcine enteral parasite of the order Coccidia, is characterized by a complex life cycle, with asexual and sexual development in the epithelium of the host gut and an environmental phase as an oocyst. All developmental stages vary greatly in their morphology and function, and therefore excrete different bioactive molecules for intercellular communication. Due to their complex development, we hypothesized that the extracellular vesicles (EVs) cargo is highly dependent on the life cycle stages from which they are released. This study aimed to characterize and compare EVs of all developmental stages of C. suis. Nanoparticle tracking analysis and microscopy were used to determine particle numbers and size distributions of stage-specific parasite EVs. Furthermore, Fourier-transform infrared spectral analysis was employed for the metabolic fingerprinting of EVs, and the lipid and protein profiles of all parasite stages were determined. Overall, the study revealed that asexual, sexual and transmissible stages of C. suis release different EVs during the parasite's life cycle. EVs of endogenous asexual and sexual stages were found to be more similar to each other than to those of the transmissible environmental stage, the oocyst. Furthermore, the ratio of fatty acids to polysaccharides and proteins changed during parasite development. In particular, proteins associated with the Apicomplexa and those involved in vesicle shedding showed changes in expression in all parasite stages. Lipid analysis showed that fatty acids were found in the same concentration through all parasite stages, whereas the amount of stereolipids, sphingolipids and glycerolipids changed between the parasite stages. In conclusion, this study, which presents the first known characterization of C. suis EVs, demonstrates a link between EVs and the respective developmental stages of the parasite, and putative functions in the parasite-parasite and host-parasite interplays.
Collapse
Affiliation(s)
- Anna Sophia Feix
- Institute of Parasitology, Department for Biological Sciences and Pathobiology, University of Veterinary Medicine Vienna, Veterinärplatz 1 A-1210 Vienna, Austria.
| | - Astrid Laimer-Digruber
- Institute of Microbiology, Department for Biological Sciences and Pathobiology, University of Veterinary Medicine, Veterinärplatz 1 A-1210 Vienna, Austria
| | - Teresa Cruz-Bustos
- Institute of Parasitology, Department for Biological Sciences and Pathobiology, University of Veterinary Medicine Vienna, Veterinärplatz 1 A-1210 Vienna, Austria
| | - Gerhard Steiner
- Department of Integrative Zoology, Faculty of Life Sciences, University of Vienna, Djerassiplatz 1 1030 Vienna, Austria
| | - Bärbel Ruttkowski
- Institute of Parasitology, Department for Biological Sciences and Pathobiology, University of Veterinary Medicine Vienna, Veterinärplatz 1 A-1210 Vienna, Austria
| | - Monika Ehling-Schulz
- Institute of Microbiology, Department for Biological Sciences and Pathobiology, University of Veterinary Medicine, Veterinärplatz 1 A-1210 Vienna, Austria
| | - Anja Joachim
- Institute of Parasitology, Department for Biological Sciences and Pathobiology, University of Veterinary Medicine Vienna, Veterinärplatz 1 A-1210 Vienna, Austria
| |
Collapse
|
20
|
Kostas JC, Brainard CS, Cristea IM. A Primer on Proteomic Characterization of Intercellular Communication in a Virus Microenvironment. Mol Cell Proteomics 2025; 24:100913. [PMID: 39862905 PMCID: PMC11889360 DOI: 10.1016/j.mcpro.2025.100913] [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: 11/17/2024] [Revised: 01/10/2025] [Accepted: 01/12/2025] [Indexed: 01/27/2025] Open
Abstract
Intercellular communication is fundamental to multicellular life and a core determinant of outcomes during viral infection, where the common goals of virus and host for persistence and replication are generally at odds. Hosts rely on encoded innate and adaptive immune responses to detect and clear viral pathogens, while viruses can exploit or disrupt these pathways and other intercellular communication processes to enhance their spread and promote pathogenesis. While virus-induced signaling can result in systemic changes to the host, striking alterations are observed within the cellular microenvironment directly surrounding a site of infection, termed the virus microenvironment (VME). Mechanisms employed by viruses to condition their VMEs are emerging and are critical for understanding the biology and pathologies of viral infections. Recent advances in experimental approaches, including proteomic methods, have enabled study of the VME in unprecedented detail. In this review article, we provide a primer on proteomic approaches used to study how viral infections alter intercellular communication, highlighting the ways in which these approaches have been implemented and the exciting biology they have uncovered. First, we consider the different molecules secreted by an infected cell, including proteins, either soluble or contained within extracellular vesicles, and metabolites. We further discuss the modalities of interactions facilitated by alteration at the cell surface of infected cells, including immunopeptide presentation and interactions with the extracellular matrix. Finally, we review spatial profiling approaches that have allowed distinguishing how specific subpopulations of cells within a VME respond to infection and alter their protein composition, discussing valuable insights these methods have offered.
Collapse
Affiliation(s)
- James C Kostas
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, USA
| | - Colter S Brainard
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, USA
| | - Ileana M Cristea
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, USA.
| |
Collapse
|
21
|
Su Z, Chao Z, Jiang V, Daniel S, Banta S. Site Directed Mutagenesis of the Cyc2 Outer Membrane Protein from Acidithiobacillus ferrooxidans Reveals a Critical Role for Bound Iron Atoms in Extracellular Electron Transfer. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2408837. [PMID: 39937138 DOI: 10.1002/smll.202408837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 12/16/2024] [Indexed: 02/13/2025]
Abstract
Extracellular electron transfer (EET) processes by metal respiratory bacteria rely on outer membrane proteins (OMPs) to exchange electrons across the insulating cell membrane. The most studied OMPs from metal reducing bacteria contain multiple sequential heme groups. However, many iron-oxidizing bacteria, including the industrial bioleaching microbe Acidithiobacillus ferrooxidans, contain monoheme OMPs and the mechanism of electron transfer through these smaller structures has not been elucidated. Computational modeling was previously used to predict two iron ion binding sites in the Cyc2 protein structure from A. ferrooxidans. To determine if these binding sites are critical for protein function, the monoheme Cyc2 OMP from A. ferrooxidans is recombinantly expressed in E. coli outer membrane vesicles (OMVs) which are then incorporated into biomimetic cell-membrane supported lipid bilayers (SLB) on electrodes to measure electron transfer. Site-directed mutagenesis is used to disrupt the putative ion binding sites predicted from modeling to elucidate the mechanism. It is confirmed that the Cyc2 protein is capable of EET without the need for soluble iron or other accessory proteins. These results confirm the critical role of bound metal ions in the A. ferrooxidans EET mechanism, and it is expected that homologous monoheme OMPs will have similar conduction pathways.
Collapse
Affiliation(s)
- Zihang Su
- Department of Chemical Engineering, Columbia University, 500 West 120th Street, New York, NY, 10027, USA
| | - Zhongmou Chao
- Robert Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, 124 Olin Hall, Ithaca, NY, 14853, USA
| | - Virginia Jiang
- Department of Chemical Engineering, Columbia University, 500 West 120th Street, New York, NY, 10027, USA
| | - Susan Daniel
- Robert Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, 124 Olin Hall, Ithaca, NY, 14853, USA
| | - Scott Banta
- Department of Chemical Engineering, Columbia University, 500 West 120th Street, New York, NY, 10027, USA
| |
Collapse
|
22
|
Xia W, Tan Y, Liu Y, Xie N, Zhu H. Prospect of extracellular vesicles in tumor immunotherapy. Front Immunol 2025; 16:1525052. [PMID: 40078996 PMCID: PMC11897508 DOI: 10.3389/fimmu.2025.1525052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Accepted: 01/28/2025] [Indexed: 03/14/2025] Open
Abstract
Extracellular vesicles (EVs), as cell-derived small vesicles, facilitate intercellular communication within the tumor microenvironment (TME) by transporting biomolecules. EVs from different sources have varied contents, demonstrating differentiated functions that can either promote or inhibit cancer progression. Thus, regulating the formation, secretion, and intake of EVs becomes a new strategy for cancer intervention. Advancements in EV isolation techniques have spurred interest in EV-based therapies, particularly for tumor immunotherapy. This review explores the multifaceted functions of EVs from various sources in tumor immunotherapy, highlighting their potential in cancer vaccines and adoptive cell therapy. Furthermore, we explore the potential of EVs as nanoparticle delivery systems in tumor immunotherapy. Finally, we discuss the current state of EVs in clinical settings and future directions, aiming to provide crucial information to advance the development and clinical application of EVs for cancer treatment.
Collapse
Affiliation(s)
- Wenbo Xia
- Department of Reproductive Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, West China Second University Hospital of Sichuan University, Chengdu, China
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Yunhan Tan
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Yongen Liu
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Na Xie
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Huili Zhu
- Department of Reproductive Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, West China Second University Hospital of Sichuan University, Chengdu, China
| |
Collapse
|
23
|
Dong X, Lin Y, Li K, Liang G, Huang X, Pan J, Wang L, Zhang D, Liu T, Wang T, Yan X, Zhang L, Li X, Qu X, Jia D, Li Y, Zhang H. Consensus statement on extracellular vesicles in liquid biopsy for advancing laboratory medicine. Clin Chem Lab Med 2025; 63:465-482. [PMID: 38896030 DOI: 10.1515/cclm-2024-0188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 05/10/2024] [Indexed: 06/21/2024]
Abstract
Extracellular vesicles (EVs) represent a diverse class of nanoscale membrane vesicles actively released by cells. These EVs can be further subdivided into categories like exosomes and microvesicles, based on their origins, sizes, and physical attributes. Significantly, disease-derived EVs have been detected in virtually all types of body fluids, providing a comprehensive molecular profile of their cellular origins. As a result, EVs are emerging as a valuable addition to liquid biopsy techniques. In this collective statement, the authors share their current perspectives on EV-related research and product development, with a shared commitment to translating this newfound knowledge into clinical applications for cancer and other diseases, particularly as disease biomarkers. The consensus within this document revolves around the overarching recognition of the merits, unresolved questions, and existing challenges surrounding EVs. This consensus manuscript is a collaborative effort led by the Committee of Exosomes, Society of Tumor Markers, Chinese anti-Cancer Association, aimed at expediting the cultivation of robust scientific and clinically applicable breakthroughs and propelling the field forward with greater swiftness and efficacy.
Collapse
Affiliation(s)
- Xingli Dong
- 558113 Central Laboratory, Department of Hematology and Oncology, Shenzhen Key Laboratory of Precision Medicine for Hematological Malignancies, Shenzhen Clinical Research Center for hematologic disease, Shenzhen University General Hospital , Shenzhen, Guangdong, China
| | - Yusheng Lin
- Department of Hematology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- Department of Thoracic Surgery, 47885 The First Affiliated Hospital of Jinan University , Guangzhou, China
- Institute of Precision Cancer Medicine and Pathology, School of Medicine
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, and MOE Key Laboratory of Tumor Molecular Biology, Jinan University, Guangzhou, China
| | - Kai Li
- Institute of Precision Cancer Medicine and Pathology, School of Medicine
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, and MOE Key Laboratory of Tumor Molecular Biology, Jinan University, Guangzhou, China
| | - Gaofeng Liang
- 74623 School of Basic Medicine and Forensic Medicine, Henan University of Science & Technology , Luoyang, China
| | - Xiaoyi Huang
- Biotherapy Center, Harbin Medical University Cancer Hospital, Heilongjiang Province, Harbin, China
- NHC Key Laboratory of Cell Transplantation, Harbin Medical University, Heilongjiang Province, Harbin, China
| | - Jingxuan Pan
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Lu Wang
- Institute of Precision Cancer Medicine and Pathology, School of Medicine
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, and MOE Key Laboratory of Tumor Molecular Biology, Jinan University, Guangzhou, China
| | - Dongmei Zhang
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, and College of Pharmacy, State Key Laboratory of Bioactive Molecules and Druggability Assessment, and MOE Key Laboratory of Tumor Molecular Biology, Jinan University, Guangzhou, China
| | - Tingjiao Liu
- Department of Oral Pathology, Shanghai Stomatological Hospital & School of Stomatology, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Fudan University, Shanghai, China
| | - Tong Wang
- 47885 MOE Key Laboratory of Tumor Molecular Biology, College of Life Science and Technology, Jinan University , Guangzhou, China
| | - Xiaomei Yan
- Department of Chemical Biology, 534787 MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, Key Laboratory for Chemical Biology of Fujian Province, Collaborative Innovation Center of Chemistry for Energy Materials, College of Chemistry and Chemical Engineering, Xiamen University , Xiamen, China
| | - Long Zhang
- 12377 MOE Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University , Hangzhou, China
| | - Xiaowu Li
- Department of Hepatobiliary Surgery, 558113 Shenzhen Key Laboratory, Shenzhen University General Hospital , Shenzhen, Guangdong, China
| | - Xiujuan Qu
- Department of Medical Oncology, 159407 The First Hospital of China Medical University , Shenyang, China
| | - Da Jia
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Department of Paediatrics, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Yong Li
- Cancer Care Centre, St George Hospital, Kogarah, NSW, Australia
- St George and Sutherland Clinical Campuses, School of Clinical Medicine, UNSW Sydney, Kensington, NSW, Australia
| | - Hao Zhang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, and MOE Key Laboratory of Tumor Molecular Biology, Jinan University, Guangzhou, China
- Institute of Precision Cancer Medicine and Pathology, and Department of Pathology, School of Medicine, Jinan University, Guangzhou, P.R. China
| |
Collapse
|
24
|
Long F, Pu X, Wang X, Ma D, Gao S, Shi J, Zhong X, Ran R, Wang L, Chen Z, Yang Y, Cannon RD, Han TL. A metabolic fingerprint of ovarian cancer: a novel diagnostic strategy employing plasma EV-based metabolomics and machine learning algorithms. J Ovarian Res 2025; 18:26. [PMID: 39940000 PMCID: PMC11823222 DOI: 10.1186/s13048-025-01590-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: 05/07/2024] [Accepted: 01/06/2025] [Indexed: 02/14/2025] Open
Abstract
Ovarian cancer (OC) is the third most common malignant tumor of women and is accompanied by an alteration of systemic metabolism. A liquid biopsy that captures and detects tumor-related biomarkers in body fluids has great potential for OC diagnosis. EVs, nanosized extracellular vesicles found in the blood, have been proposed as promising biomarkers for liquid biopsies. In this study we recruited 37 OC patients, 22 benign ovarian tumor (BE) patients, and 46 clinically healthy control patients (CON). Plasma EVs were purified from blood samples and sensitive thermal separation probe-based mass spectrometry analysis using a global untargeted metabolic profiling strategy was employed to characterize the metabolite fingerprints. Uniform manifold approximation and projection (UMAP) analysis demonstrated a distinct separation of EVs among the three groups. We screened for diagnostic biomarkers from plasma EV metabolites using seven machine learning algorithms, including artificial neural network (ANN), decision tree (DT), K nearest neighbor (KNN), logistics regression (LR), Naïve Bayes (NB), random forest (RF), and support vector machine (SVM). For the OC-CON comparison, the highest AUC values were found for RF (0.91), ANN (0.90) and NB (0.90), with the F1-scores of 0.88, 0.83, and 0.76 respectively. For the OC-BE comparison, SVM (0.94), RF (0.86), and KNN (0.86) gave the highest AUCs, with F1-scores of 0.80, 0.80, and 0.91 respectively. A total of 19 and 158 metabolic features exhibited significant differences (FC = 1.5, q < 0.01) in the OC vs BE and OC vs CON comparisons, respectively. Notably, the quantities of 9-octadecenamide and 1,4-methanobenzocyclodecene were significantly elevated, while maltol showed a significant reduction in the OC group compared to the BE group. When comparing the OC group to the CON group, the concentrations of 4-amino-furazan-3-carboxylic acid 2-hydroxy-4-methoxybenzaldehyde, N-phenylethyl, and 4-morpholineethanamine were significantly elevated, while the remaining metabolites, including hydrazine and pyridine sulfonamide, were reduced, in the OC group. The metabolites showing different abundancies are associated with cancer-related mutations, immune responses, and metabolic reprogramming. We demonstrate that the RF algorithm, combined with sensitive thermal separation probe-based mass spectrometry analysis of plasma EVs, can effectively identify OC patients with good accuracy. Thus, our study has shortlisted a set of potential biomarkers in plasma EVs, and the proposed approach could serve as a routine prescreening tool for ovarian cancer.
Collapse
Affiliation(s)
- Fei Long
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Biomedical Engineering, Chongqing Medical University, Chongqing, China
| | - XingYu Pu
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Biomedical Engineering, Chongqing Medical University, Chongqing, China
| | - Xin Wang
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Biomedical Engineering, Chongqing Medical University, Chongqing, China
| | - DongXue Ma
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Biomedical Engineering, Chongqing Medical University, Chongqing, China
| | - ShanHu Gao
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Biomedical Engineering, Chongqing Medical University, Chongqing, China
| | - Jun Shi
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Biomedical Engineering, Chongqing Medical University, Chongqing, China
| | - XiaoCui Zhong
- Department of Obstetrics and Gynaecology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Rui Ran
- Department of Obstetrics and Gynaecology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - LianLian Wang
- Department of Reproductive Center, The First Affiliated Hospital of Chongqing, Medical University, Chongqing, China
| | - Zhu Chen
- Department of Obstetrics and Gynecology, Second Affiliated HospitalArmy Medical University, Chongqing, China
| | - Yang Yang
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Richard D Cannon
- Department of Oral Sciences, Faculty of Dentistry, Sir John Walsh Research Institute, University of Otago, Dunedin, New Zealand
| | - Ting-Li Han
- Department of Obstetrics and Gynaecology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China.
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, China.
- Chongqing Key Laboratory of Biomedical Engineering, Chongqing Medical University, Chongqing, China.
| |
Collapse
|
25
|
Chen J, Madhiyan M, Moor KJ, Chen H, Shuai D. Kinetics and Mechanisms of Solar UVB Disinfection of Vesicle-Cloaked Murine Norovirus Clusters and Free Noroviruses. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2025; 59:2461-2472. [PMID: 39893675 DOI: 10.1021/acs.est.4c12583] [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: 02/04/2025]
Abstract
Human norovirus, a major global cause of gastroenteritis, forms vesicle-cloaked virus clusters (known as viral vesicles), showing increased infectivity and persistence in aquatic environments. We investigated UVB disinfection, a key mechanism of solar disinfection commonly employed in developing countries, targeting murine norovirus vesicles and free murine noroviruses as surrogates for human noroviruses. At low viral concentrations of 109 gene copies per liter, viral infectivity loss as quantified by the integrated cell culture-reverse transcription-quantitative polymerase chain reaction (ICC-RT-qPCR) indicated that vesicles were 1.51 to 1.73 times more resistant to disinfection compared to free viruses. Virus inactivation was primarily due to protein damage as quantified by bicinchoninic acid and Western blot assays, and the damage of virus binding to host cells as quantified by RT-qPCR. Molecular simulations predicted that the oxidation of a tyrosine residue in the viral protein 1 prohibited binding. UVB irradiation of viral/vesicle proteins resulted in 1O2 formation as quantified by time-resolved phosphorescence, and for the first time, endogenous 1O2 was confirmed to contribute to virus inactivation by UVB. Our study recognizes the limitation of UVB disinfection of viral vesicles particularly in solar wastewater treatment and advocates for enhanced disinfection strategies to protect public health.
Collapse
Affiliation(s)
- Jiahao Chen
- Department of Civil and Environmental Engineering, The George Washington University, Washington, District of Columbia 20052, United States
| | - Monika Madhiyan
- Utah Water Research Laboratory, Department of Civil and Environmental Engineering, Utah State University, Logan, Utah 84322, United States
| | - Kyle J Moor
- Utah Water Research Laboratory, Department of Civil and Environmental Engineering, Utah State University, Logan, Utah 84322, United States
| | - Hanning Chen
- Texas Advanced Computing Center, The University of Texas at Austin, Austin, Texas 78758, United States
| | - Danmeng Shuai
- Department of Civil and Environmental Engineering, The George Washington University, Washington, District of Columbia 20052, United States
| |
Collapse
|
26
|
Mohamed AH, Abaza T, Youssef YA, Rady M, Fahmy SA, Kamel R, Hamdi N, Efthimiado E, Braoudaki M, Youness RA. Extracellular vesicles: from intracellular trafficking molecules to fully fortified delivery vehicles for cancer therapeutics. NANOSCALE ADVANCES 2025; 7:934-962. [PMID: 39823046 PMCID: PMC11733735 DOI: 10.1039/d4na00393d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 12/22/2024] [Indexed: 01/19/2025]
Abstract
Extracellular vesicles (EVs) are emerging as viable tools in cancer treatment due to their ability to carry a wide range of theranostic activities. This review summarizes different forms of EVs such as exosomes, microvesicles, apoptotic bodies, and oncosomes. It also sheds the light onto isolation methodologies, characterization techniques and therapeutic applications of all discussed EVs. Evidence indicates that EVs are particularly effective in delivering chemotherapeutic medications, and immunomodulatory agents. However, the advancement of EV-based therapies into clinical practice is hindered by challenges including EVs heterogeneity, cargo loading efficiency, and in vivo stability. Overall, EVs have the potential to change cancer therapeutic paradigms. Continued research and development activities are critical for improving EV-based medications and increasing their therapeutic impact.
Collapse
Affiliation(s)
- Adham H Mohamed
- Department of Chemistry, Faculty of Science, Cairo University 12613 Giza Egypt
| | - Tasneem Abaza
- Biotechnology and Biomolecular Chemistry Program, Faculty of Science, Cairo University 12613 Giza Egypt
- Université Paris-Saclay, Université d'Evry Val D'Essonne 91000 Évry-Courcouronnes Île-de-France France
| | - Yomna A Youssef
- Department of Physiology, Faculty of Physical Therapy, German International University (GIU) 11835 Cairo Egypt
- Molecular Biology and Biochemistry Department, Faculty of Biotechnology, German International University (GIU) 11835 Cairo Egypt
| | - Mona Rady
- Microbiology, Immunology and Biotechnology Department, Faculty of Pharmacy and Biotechnology, German University in Cairo (GUC) 11835 Cairo Egypt
- Faculty of Biotechnology, German International University New Administrative Capital 11835 Cairo Egypt
| | - Sherif Ashraf Fahmy
- Department of Pharmaceutics and Biopharmaceutics, University of Marburg Robert-Koch-Str. 4 35037 Marburg Germany
| | - Rabab Kamel
- Pharmaceutical Technology Department, National Research Centre 12622 Cairo Egypt
| | - Nabila Hamdi
- Pharmacology and Toxicology Department, Faculty of Pharmacy and Biotechnology, German University in Cairo (GUC) 11835 Cairo Egypt
| | - Eleni Efthimiado
- Inorganic Chemistry Laboratory, Chemistry Department, National and Kapodistrian University of Athens Athens Greece
| | - Maria Braoudaki
- Department of Clinical, Pharmaceutical, and Biological Science, School of Life and Medical Sciences, University of Hertfordshire Hatfield AL10 9AB UK
| | - Rana A Youness
- Molecular Biology and Biochemistry Department, Faculty of Biotechnology, German International University (GIU) 11835 Cairo Egypt
| |
Collapse
|
27
|
Lu Y, Chen T, Lin H, Chen Y, Lin Y, Le D, Huang Y, Wang AH, Lee C, Ling T. Small Extracellular Vesicles Engineered Using Click Chemistry to Express Chimeric Antigen Receptors Show Enhanced Efficacy in Acute Liver Failure. J Extracell Vesicles 2025; 14:e70044. [PMID: 39901768 PMCID: PMC11791321 DOI: 10.1002/jev2.70044] [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: 07/02/2024] [Revised: 01/06/2025] [Accepted: 01/10/2025] [Indexed: 02/05/2025] Open
Abstract
Acetaminophen (APAP) overdose can cause severe liver injury and life-threatening conditions that may lead to multiple organ failure without proper treatment. N-acetylcysteine (NAC) is the accepted and prescribed treatment for detoxification in cases of APAP overdose. Nonetheless, in acute liver failure (ALF), particularly when the ingestion is substantial, NAC may not fully restore liver function. NAC administration in ALF has limitations and potential adverse effects, including nausea, vomiting, diarrhoea, flatus, gastroesophageal reflux, and anaphylactoid reactions. Mesenchymal stromal cell (MSC)-based therapies using paracrine activity show promise for treating ALF, with preclinical studies demonstrating improvement. Recently, MSC-derived extracellular vesicles (EVs) have emerged as a new therapeutic option for liver injury. MSC-derived EVs can contain various therapeutic cargos depending on the cell of origin, participate in physiological processes, and respond to abnormalities. However, most therapeutic EVs lack a distinct orientation upon entering the body, resulting in a lack of targeting specificity. Therefore, enhancing the precision of natural EV delivery systems is urgently needed. Thus, we developed an advanced targeting technique to deliver modified EVs within the body. Our strategy aims to employ bioorthogonal click chemistry to attach a targeting molecule to the surface of small extracellular vesicles (sEVs), creating exogenous chimeric antigen receptor-modified sEVs (CAR-sEVs) for the treatment. First, we engineered azido-modified sEVs (N3-sEVs) through metabolic glycoengineering by treating MSCs with the azide-containing monosaccharide N-azidoacetyl-mannosamine (Ac4ManNAz). Next, we conjugated N3-sEVs with a dibenzocyclooctyne (DBCO)-tagged single-chain variable fragment (DBCO-scFv) that targets the asialoglycoprotein receptor (ASGR1), thus producing CAR-sEVs for precise liver targeting. The efficacy of CAR-sEV therapy in ALF models by targeting ASGR1 was validated. MSC-derived CAR-sEVs reduced serum liver enzymes, mitigated liver damage, and promoted hepatocyte proliferation in APAP-induced injury. Overall, CAR-sEVs exhibited enhanced hepatocyte specificity and efficacy in ameliorating liver injury, highlighting the significant advancements achievable with cell-free targeted therapy.
Collapse
Affiliation(s)
- Yen‐Ting Lu
- Graduate Institute of PharmacologyNational Taiwan University College of MedicineTaipeiTaiwan
| | - Tzu‐Yu Chen
- Graduate Institute of PharmacologyNational Taiwan University College of MedicineTaipeiTaiwan
| | - Hsin‐Hung Lin
- Graduate Institute of PharmacologyNational Taiwan University College of MedicineTaipeiTaiwan
- MediDiamond Inc.TaipeiTaiwan
| | - Ya‐Wen Chen
- The Ph.D. Program for Translational Medicine, College of Medical Science and TechnologyTaipei Medical UniversityTaipeiTaiwan
| | - Yu‐Xiu Lin
- Graduate Institute of PharmacologyNational Taiwan University College of MedicineTaipeiTaiwan
| | - Duy‑Cuong Le
- International Ph.D. Program in Cell Therapy and Regenerative Medicine, College of MedicineTaipei Medical UniversityTaipeiTaiwan
- Vinmec Hi‐Tech CenterVinmec Healthcare SystemHanoiVietnam
- International Ph.D. Program for Translational Science, College of Medical Science and TechnologyTaipei Medical UniversityTaipeiTaiwan
| | - Yen‐Hua Huang
- International Ph.D. Program in Cell Therapy and Regenerative Medicine, College of MedicineTaipei Medical UniversityTaipeiTaiwan
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of MedicineTaipei Medical UniversityTaipeiTaiwan
- Graduate Institute of Medical Sciences, College of MedicineTaipei Medical UniversityTaipeiTaiwan
- TMU Research Center of Cell Therapy and Regeneration MedicineTaipei Medical UniversityTaipeiTaiwan
| | - Andrew H.‐J. Wang
- The Ph.D. Program for Translational Medicine, College of Medical Science and TechnologyTaipei Medical UniversityTaipeiTaiwan
| | - Cheng‐Chung Lee
- The Ph.D. Program for Translational Medicine, College of Medical Science and TechnologyTaipei Medical UniversityTaipeiTaiwan
- International Ph.D. Program for Translational Science, College of Medical Science and TechnologyTaipei Medical UniversityTaipeiTaiwan
| | - Thai‐Yen Ling
- Graduate Institute of PharmacologyNational Taiwan University College of MedicineTaipeiTaiwan
| |
Collapse
|
28
|
Sung SE, Seo MS, Park WT, Lim YJ, Park S, Lee GW. Extracellular vesicles: their challenges and benefits as potential biomarkers for musculoskeletal disorders. J Int Med Res 2025; 53:3000605251317476. [PMID: 39973226 PMCID: PMC11840854 DOI: 10.1177/03000605251317476] [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: 07/09/2024] [Accepted: 01/10/2025] [Indexed: 02/21/2025] Open
Abstract
Early diagnosis and timely management are critical for determining disease outcomes and prognoses. To date, certain methods for developing disease-specific biomarkers have been reported; however, strategies for musculoskeletal disease-specific biomarker development have rarely been studied. Recent studies have highlighted the potential application of extracellular vesicles (EVs) as disease-specific biomarkers. EVs encapsulate proteins, lipids, messenger RNAs, and microRNAs derived from their cellular origin; these constituents remain stable within the EVs and can traverse the blood-brain barrier. Because of these distinctive characteristics, EVs have been actively investigated as diagnostic tools for various conditions, including cancer, inflammatory diseases, and musculoskeletal disorders. Although EVs have many advantages for biomarker development, they have not yet been fully researched in the context of musculoskeletal pathologies. The current review aimed to highlight the potential of EVs in the development of disease-specific biomarkers, summarize the processes of EV biomarkers, and discuss current limitations and future perspectives of EVs as biomarkers.
Collapse
Affiliation(s)
- Soo-Eun Sung
- Preclinical Research Center, Daegu-Gyeongbuk Medical Innovation Foundation (K-MEDI hub), Daegu, Republic of Korea
| | - Min-Soo Seo
- Department of Veterinary Tissue Engineering, College of Veterinary Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Wook-Tae Park
- Department of Orthopedic Surgery, Yeungnam University College of Medicine, Yeungnam University Medical Center, Daegu, Republic of Korea
| | - Young-Ju Lim
- Department of Orthopedic Surgery, Yeungnam University College of Medicine, Yeungnam University Medical Center, Daegu, Republic of Korea
| | - Sangbum Park
- Department of Cell and Molecular Biology, The Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, USA
| | - Gun Woo Lee
- Department of Orthopedic Surgery, Yeungnam University College of Medicine, Yeungnam University Medical Center, Daegu, Republic of Korea
- Department of Cell and Molecular Biology, The Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, USA
| |
Collapse
|
29
|
Wood NA, Gopinath T, Shin K, Marassi FM. In situ NMR reveals a pH sensor motif in an outer membrane protein that drives bacterial vesicle production. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.21.634179. [PMID: 39896486 PMCID: PMC11785132 DOI: 10.1101/2025.01.21.634179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
The outer membrane vesicles (OMVs) produced by diderm bacteria have important roles in cell envelope homeostasis, secretion, interbacterial communication, and pathogenesis. The facultative intracellular pathogen Salmonella enterica Typhimurium (STm) activates OMV biogenesis inside the acidic vacuoles of host cells by upregulating the expression of the outer membrane (OM) protein PagC, one of the most robustly activated genes in a host environment. Here, we used solid-state nuclear magnetic resonance (NMR) and electron microscopy (EM), with native bacterial OMVs, to demonstrate that three histidines, essential for the OMV biogenic function of PagC, constitute a key pH-sensing motif. The NMR spectra of PagC in OMVs show that they become protonated around pH 6, and His protonation is associated with specific perturbations of select regions of PagC. The use of bacterial OMVs is an essential aspect of this work enabling NMR structural studies in the context of the physiological environment. PagC expression upregulates OMV production in E. coli, replicating its function in STm. Moreover, the presence of PagC drives a striking aggregation of OMVs and increases bacterial cell pellicle formation at acidic pH, pointing to a potential role as an adhesin active in biofilm formation. The data provide experimental evidence for a pH-dependent mechanism of OMV biogenesis and aggregation driven by an outer membrane protein.
Collapse
Affiliation(s)
- Nicholas A Wood
- Department of Biophysics, Medical College of Wisconsin 8701 Watertown Plank Road, Milwaukee, WI 53226-3548 USA
| | - Tata Gopinath
- Department of Biophysics, Medical College of Wisconsin 8701 Watertown Plank Road, Milwaukee, WI 53226-3548 USA
| | - Kyungsoo Shin
- Department of Biophysics, Medical College of Wisconsin 8701 Watertown Plank Road, Milwaukee, WI 53226-3548 USA
| | - Francesca M. Marassi
- Department of Biophysics, Medical College of Wisconsin 8701 Watertown Plank Road, Milwaukee, WI 53226-3548 USA
| |
Collapse
|
30
|
Khan A, Khan H, He N, Li Z, Alyahya HK, Bin Jardan YA. Colorimetric aptasensor coupled with a deep-learning-powered smartphone app for programmed death ligand-1 expressing extracellular vesicles. Front Immunol 2025; 15:1479403. [PMID: 39916963 PMCID: PMC11798968 DOI: 10.3389/fimmu.2024.1479403] [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: 08/13/2024] [Accepted: 11/21/2024] [Indexed: 02/09/2025] Open
Abstract
Lung cancer is a devastating public health threat and a leading cause of cancer-related deaths. Therefore, it is imperative to develop sophisticated techniques for the non-invasive detection of lung cancer. Extracellular vesicles expressing programmed death ligand-1 (PD-L1) markers (PD-L1@EVs) in the blood are reported to be indicative of lung cancer and response to immunotherapy. Our approach is the development of a colorimetric aptasensor by combining the rapid capturing efficiency of (Fe3O4)-SiO2-TiO2 for EV isolation with PD-L1 aptamer-triggered enzyme-linked hybridization chain reaction (HCR) for signal amplification. The numerous HRPs catalyze their substrate dopamine (colorless) into polydopamine (blackish brown). Change in chromaticity directly correlates with the concentration of PD-L1@EVs in the sample. The colorimetric aptasensor was able to detect PD-L1@EVs at concentrations as low as 3.6×102 EVs/mL with a wide linear range from 103 to 1010 EVs/mL with high specificity and successfully detected lung cancer patients' serum from healthy volunteers' serum. To transform the qualitative colorimetric approach into a quantitative operation, we developed an intelligent convolutional neural network (CNN)-powered quantitative analyzer for chromaticity in the form of a smartphone app named ExoP, thereby achieving the intelligent analysis of chromaticity with minimal user intervention or additional hardware attachments for the sensitive and specific quantification of PD-L1@EVs. This combined approach offers a simple, sensitive, and specific tool for lung cancer detection using PD-L1@EVs. The addition of a CNN-powered smartphone app further eliminates the need for specialized equipment, making the colorimetric aptasensor more accessible for low-resource settings.
Collapse
Affiliation(s)
- Adeel Khan
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
- Department of Biotechnology, University of Science and Technology, Bannu, Pakistan
| | - Haroon Khan
- Neuroscience and Neuroengineering Research Center, Med-X Research Institute, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Nongyue He
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
| | - Zhiyang Li
- Department of Clinical Laboratory, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Heba Khalil Alyahya
- Department of Exercise Physiology, College of Sport Science and Physical Activity, King Saud University, Riyadh, Saudi Arabia
| | - Yousef A. Bin Jardan
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
31
|
Dogan AB, Marsh SR, Tschetter RJ, E Beard C, Amin MR, Jane Jourdan L, Gourdie RG. Stabilizing milk-derived extracellular vesicles (mEVs) through lyophilization: a novel trehalose and tryptophan formulation for maintaining structure and Bioactivity during long-term storage. J Biol Eng 2025; 19:4. [PMID: 39806456 PMCID: PMC11727230 DOI: 10.1186/s13036-024-00470-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: 08/26/2024] [Accepted: 11/26/2024] [Indexed: 01/16/2025] Open
Abstract
Extracellular vesicles (EVs) are widely investigated for their implications in cell-cell signaling, immune modulation, disease pathogenesis, cancer, regenerative medicine, and as a potential drug delivery vector. However, maintaining integrity and bioactivity of EVs between Good Manufacturing Practice separation/filtration and end-user application remains a consistent bottleneck towards commercialization. Milk-derived extracellular vesicles (mEVs), separated from bovine milk, could provide a relatively low-cost, scalable platform for large-scale mEV production; however, the reliance on cold supply chain for storage remains a logistical and financial burden for biologics that are unstable at room temperature. Herein, we aim to characterize and engineer a freeze-dried, mEV formulation that can be stored at room temperature without sacrificing structure/bioactivity and can be reconstituted before delivery. In addition to undertaking established mEV assays of structure and function on our preparations, we introduce a novel, efficient, high throughput assay of mEV bioactivity based on Electric Cell Substrate Impedance Sensing (ECIS) in Human dermal fibroblast monolayers. By adding appropriate excipients, such as trehalose and tryptophan, we describe a protective formulation that preserves mEV bioactivity during long-term, room temperature storage. Our identification of the efficacy of tryptophan as a novel additive to mEV lyophilization solutions could represent a significant advancement in stabilizing small extracellular vesicles outside of cold storage conditions.
Collapse
Affiliation(s)
- Alan B Dogan
- Virginia Tech Carilion School of Medicine, Roanoke, VA, 24016, USA
| | - Spencer R Marsh
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA, 24016, USA
- Center for Vascular and Heart Research, Virginia Tech, Roanoke, VA, 24016, USA
| | - Rachel J Tschetter
- Materials Science and Engineering, Virginia Tech, Blacksburg, VA, 24061, USA
| | - Claire E Beard
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA, 24016, USA
| | - Md R Amin
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA, 24016, USA
- Translational Biology, Medicine, and Health graduate program at Virginia Tech, Roanoke, VA, 24016, USA
| | - L Jane Jourdan
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA, 24016, USA
- Center for Vascular and Heart Research, Virginia Tech, Roanoke, VA, 24016, USA
| | - Robert G Gourdie
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA, 24016, USA.
- Center for Vascular and Heart Research, Virginia Tech, Roanoke, VA, 24016, USA.
- Department of Biomedical Engineering and Mechanics, Virginia Tech, Blacksburg, VA, 24061, USA.
- Department of Emergency Medicine, Virginia Tech Carilion School of Medicine, Virginia Tech, Roanoke, VA, 24016, USA.
- Faculty of Health Science, Virginia Tech, Blacksburg, VA, 24061, USA.
| |
Collapse
|
32
|
Guo J, Huang Z, Wang Q, Wang M, Ming Y, Chen W, Huang Y, Tang Z, Huang M, Liu H, Jia B. Opportunities and challenges of bacterial extracellular vesicles in regenerative medicine. J Nanobiotechnology 2025; 23:4. [PMID: 39754127 PMCID: PMC11697683 DOI: 10.1186/s12951-024-02935-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 10/16/2024] [Indexed: 01/07/2025] Open
Abstract
Extracellular vesicles (EVs) are membrane-bound vesicles that are shed or secreted from the cell membrane and enveloped by a lipid bilayer. They possess stability, low immunogenicity, and non-cytotoxicity, exhibiting extensive prospects in regenerative medicine (RM). However, natural EVs pose challenges, such as insufficient targeting capabilities, potential biosafety concerns, and limited acquisition pathways. Although engineered EVs demonstrate excellent therapeutic efficacy, challenges such as low production yield and the complexity of engineering modifications constrain their further clinical applications. Bacteria have advantages such as rapid proliferation, diverse gene editing methods, mature cultivation techniques, and relatively easy preparation of bacterial EVs (BEVs), which can be used to effectively address the challenges currently encountered in the field of EVs. This review provides a description of the biogenesis and pathophysiological functions of BEVs, and strategies for optimizing BEVs preparation to attain efficiency and safety are discussed. An analysis of natural characteristics of BEVs is also conducted to explore how to leverage their advantages or mitigate their limitations, thereby overcoming constraints on the application of BEVs in RM. In summary, engineered BEVs possess characteristics such as high production yield, excellent stability, and high drug-delivering capabilities, laying the foundation for their application in RM.
Collapse
Affiliation(s)
- Jiming Guo
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China
| | - Zhijie Huang
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China
| | - Qinjing Wang
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China
| | - Min Wang
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China
| | - Yue Ming
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China
| | - Weixing Chen
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China
| | - Yisheng Huang
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China
| | - Zhengming Tang
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China
| | - Mingshu Huang
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China
| | - Hongyu Liu
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China
| | - Bo Jia
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
33
|
Vitucci ECM, Carberry CK, Payton A, Herring LE, Mordant AL, Kim YH, Gilmour MI, McCullough SD, Rager JE. Wildfire-relevant woodsmoke and extracellular vesicles (EVs): Alterations in EV proteomic signatures involved in extracellular matrix degradation and tissue injury in airway organotypic models. ENVIRONMENTAL RESEARCH 2025; 264:120395. [PMID: 39571711 DOI: 10.1016/j.envres.2024.120395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 10/16/2024] [Accepted: 11/18/2024] [Indexed: 12/01/2024]
Abstract
Wildfires adversely impact air quality and public health worldwide. Exposures to wildfire smoke are linked to adverse health outcomes, including cardiopulmonary diseases. Critical research gaps remain surrounding the underlying biological pathways leading to wildfire-induced health effects. The regulation of intercellular communication and downstream toxicity driven by extracellular vesicles (EVs) is an important, understudied biological mechanism. This study investigated EVs following a wildfire smoke-relevant in vitro exposure. We hypothesized that woodsmoke (WS) would alter the proteomic content of EVs secreted in organotypic in vitro airway models. Exposures were carried out using a tri-culture model of alveolar epithelial cells, fibroblasts, and endothelial cells and a simplified co-culture model of alveolar epithelial cells and fibroblasts to inform responses across different cell populations. Epithelial cells were exposed to WS condensate and EVs were isolated from basolateral conditioned medium following 24 h exposure. WS exposure did not influence EV particle characteristics, and it moderately increased EV count. Exposure caused the differential loading of 25 and 35 proteins within EVs collected from the tri- and co-culture model, respectively. EV proteins involved in extracellular matrix degradation and wound healing were consistently modulated across both models. However, distinct proteins involved in the wound healing pathway were altered between models, suggesting unique but concerted efforts across cell types to communicate in response to injury. These findings demonstrate that a wildfire-relevant exposure alters the EV proteome and suggest an impact on EV-mediated intercellular communication. Overall, results demonstrate the viability of organotypic approaches in evaluating EVs to investigate exposure-induced biomarkers and underlying mechanisms. Findings also highlight the impact of differences in the biological complexity of in vitro models used to evaluate the effects of inhaled toxicants.
Collapse
Affiliation(s)
- Eva C M Vitucci
- Interdisciplinary Faculty of Toxicology, School of Public Health, Texas A&M University, College Station, TX, USA; Curriculum in Toxicology & Environmental Medicine, School of Medicine, University of North Carolina, Chapel Hill, NC, USA; The Center for Environmental Medicine, Asthma and Lung Biology, School of Medicine, The University of North Carolina, Chapel Hill, NC, USA
| | - Celeste K Carberry
- Department of Environmental Sciences and Engineering, Gillings School of Global Public Health, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; The Institute for Environmental Health Solutions, Gillings School of Global Public Health, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Alexis Payton
- The Center for Environmental Medicine, Asthma and Lung Biology, School of Medicine, The University of North Carolina, Chapel Hill, NC, USA; Department of Environmental Sciences and Engineering, Gillings School of Global Public Health, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Laura E Herring
- UNC Proteomics Core Facility, Department of Pharmacology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Angie L Mordant
- UNC Proteomics Core Facility, Department of Pharmacology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Yong Ho Kim
- Public Health and Integrated Toxicology Division, Center for Public Health and Environmental Assessment, U.S. Environmental Protection Agency, Research Triangle Park, NC, USA
| | - M Ian Gilmour
- Public Health and Integrated Toxicology Division, Center for Public Health and Environmental Assessment, U.S. Environmental Protection Agency, Research Triangle Park, NC, USA
| | - Shaun D McCullough
- Curriculum in Toxicology & Environmental Medicine, School of Medicine, University of North Carolina, Chapel Hill, NC, USA; Exposure and Protection Group, Technology Advancement and Commercialization Unit, Research Triangle Institute International, Durham, NC 27709, USA; Public Health and Integrated Toxicology Division, Center for Public Health and Environmental Assessment, U.S. Environmental Protection Agency, Chapel Hill, NC, USA.
| | - Julia E Rager
- Curriculum in Toxicology & Environmental Medicine, School of Medicine, University of North Carolina, Chapel Hill, NC, USA; The Center for Environmental Medicine, Asthma and Lung Biology, School of Medicine, The University of North Carolina, Chapel Hill, NC, USA; Department of Environmental Sciences and Engineering, Gillings School of Global Public Health, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; The Institute for Environmental Health Solutions, Gillings School of Global Public Health, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
34
|
Cai J, Liu Y. Isolation and Characterization of Exosomes from Ocular Samples. Methods Mol Biol 2025; 2858:63-75. [PMID: 39433667 PMCID: PMC11574743 DOI: 10.1007/978-1-0716-4140-8_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] [Indexed: 10/23/2024]
Abstract
Exosomes are microsize vesicles secreted by nearly all cells to the extracellular space. The vesicles transport cell signaling and communicate with other cells. Ultracentrifugation is the standard method to isolate exosomes from culture media or body fluid. Without ultracentrifuge, exosomes can be precipitated by polyethylene glycol or separated by size exclusion chromatography. After isolation, nanoparticle tracking analysis can help to estimate the size and concentration of exosome samples. Transmission electron microscopy can directly show the size and morphology of exosomes. Moreover, the sample should be characterized by the expression of several exosome biomarker proteins. Exosomal contents such as proteins and miRNAs could be profiled using appropriate technologies.
Collapse
Affiliation(s)
- Jingwen Cai
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Yutao Liu
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, USA.
| |
Collapse
|
35
|
Ghodsi M, Cloos A, Lotens A, De Bueger M, Van Der Smissen P, Henriet P, Cellier N, Pierreux CE, Najdovski T, Tyteca D. Development of an easy non-destructive particle isolation protocol for quality control of red blood cell concentrates. JOURNAL OF EXTRACELLULAR BIOLOGY 2025; 4:e70028. [PMID: 39830833 PMCID: PMC11739896 DOI: 10.1002/jex2.70028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 11/29/2024] [Accepted: 12/10/2024] [Indexed: 01/22/2025]
Abstract
The extracellular vesicle release in red blood cell concentrates reflects progressive accumulation of storage lesions and could represent a new measure to be implemented routinely in blood centres in addition to haemolysis. Nevertheless, there is currently no standardized isolation protocol. In a previous publication, we developed a reproducible ultracentrifugation-based protocol (20,000 × g protocol) that allows to classify red blood cell concentrates into three cohorts according to their vesiculation level. Since this protocol was not adapted to meet routine requirements, the goal of this study was to develop an easier method based on low-speed centrifugation (2,000 × g protocol) and limited red blood cell concentrate volumes to match with a non-destructive sampling from the quality control sampling tubing. Despite the presence of contaminants, mainly in the form of albumin and lipoproteins, the material isolated with the 2,000 × g protocol contained red blood cell-derived vesicular structures. It was reproducible, could predict the number of extracellular vesicles obtained with the 20,000 × g protocol and better discriminated between the three vesiculation cohorts than haemolysis at the legal expiry date of 6 weeks. However, by decreasing red blood cell concentrate volumes to fit with the volume in the quality control tubing, particle yield was highly reduced. Therefore, centrifugation time and relative centrifugal force were adapted (1,000 × g protocol), allowing for the recovery of a similar particle number and composition between small and large volumes sampled from the main unit, in different vesiculation cohorts over time. A similar observation was made with the 1,000 × g protocol between small volumes sampled from the quality control tubing and the mother-bag. In conclusion, our study paves the way for the use of the 2,000 × g protocol (adapted to a 1,000 × g protocol with the quality control sampling tubing) for particle measurement in blood centres.
Collapse
Affiliation(s)
- Marine Ghodsi
- Cell Biology Unit & Platform for Imaging Cells and Tissues, de Duve InstituteUCLouvainBrusselsBelgium
| | - Anne‐Sophie Cloos
- Cell Biology Unit & Platform for Imaging Cells and Tissues, de Duve InstituteUCLouvainBrusselsBelgium
| | - Anaïs Lotens
- Service du SangCroix‐Rouge de BelgiqueSuarléeBelgium
| | - Marine De Bueger
- Cell Biology Unit & Platform for Imaging Cells and Tissues, de Duve InstituteUCLouvainBrusselsBelgium
| | - Patrick Van Der Smissen
- Cell Biology Unit & Platform for Imaging Cells and Tissues, de Duve InstituteUCLouvainBrusselsBelgium
| | - Patrick Henriet
- Cell Biology Unit & Platform for Imaging Cells and Tissues, de Duve InstituteUCLouvainBrusselsBelgium
| | | | - Christophe E. Pierreux
- Cell Biology Unit & Platform for Imaging Cells and Tissues, de Duve InstituteUCLouvainBrusselsBelgium
| | | | - Donatienne Tyteca
- Cell Biology Unit & Platform for Imaging Cells and Tissues, de Duve InstituteUCLouvainBrusselsBelgium
| |
Collapse
|
36
|
Carney RP, Mizenko RR, Bozkurt BT, Lowe N, Henson T, Arizzi A, Wang A, Tan C, George SC. Harnessing extracellular vesicle heterogeneity for diagnostic and therapeutic applications. NATURE NANOTECHNOLOGY 2025; 20:14-25. [PMID: 39468355 PMCID: PMC11781840 DOI: 10.1038/s41565-024-01774-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 07/11/2024] [Indexed: 10/30/2024]
Abstract
Extracellular vesicles (EVs) are diverse nanoparticles with large heterogeneity in size and molecular composition. Although this heterogeneity provides high diagnostic value for liquid biopsy and confers many exploitable functions for therapeutic applications in cancer detection, wound healing and neurodegenerative and cardiovascular diseases, it has also impeded their clinical translation-hence heterogeneity acts as a double-edged sword. Here we review the impact of subpopulation heterogeneity on EV function and identify key cornerstones for addressing heterogeneity in the context of modern analytical platforms with single-particle resolution. We outline concrete steps towards the identification of key active biomolecules that determine EV mechanisms of action across different EV subtypes. We describe how such knowledge could accelerate EV-based therapies and engineering approaches for mimetic artificial nanovesicle formulations. This approach blunts one edge of the sword, leaving only a single razor-sharp edge on which EV heterogeneity can be exploited for therapeutic applications across many diseases.
Collapse
Affiliation(s)
- Randy P Carney
- Department of Biomedical Engineering, University of California, Davis, Davis, CA, USA.
| | - Rachel R Mizenko
- Department of Biomedical Engineering, University of California, Davis, Davis, CA, USA
| | - Batuhan T Bozkurt
- Department of Biomedical Engineering, University of California, Davis, Davis, CA, USA
| | - Neona Lowe
- Department of Biomedical Engineering, University of California, Davis, Davis, CA, USA
| | - Tanner Henson
- Department of Biomedical Engineering, University of California, Davis, Davis, CA, USA
- Center for Surgical Bioengineering, Department of Surgery, University of California, Davis, School of Medicine, Sacramento, CA, USA
| | - Alessandra Arizzi
- Department of Biomedical Engineering, University of California, Davis, Davis, CA, USA
| | - Aijun Wang
- Department of Biomedical Engineering, University of California, Davis, Davis, CA, USA
- Center for Surgical Bioengineering, Department of Surgery, University of California, Davis, School of Medicine, Sacramento, CA, USA
| | - Cheemeng Tan
- Department of Biomedical Engineering, University of California, Davis, Davis, CA, USA
| | - Steven C George
- Department of Biomedical Engineering, University of California, Davis, Davis, CA, USA.
- Center for Surgical Bioengineering, Department of Surgery, University of California, Davis, School of Medicine, Sacramento, CA, USA.
| |
Collapse
|
37
|
Szigyártó IC, Singh P, Sonallya T, Románszki L, Mihály J, Wiener Z, Bebesi T, Mészáros G, Keresztes Z, Thompson M, Varga Z, Beke-Somfai T. ATR-Infrared Spectroscopy and Acoustic Sensing in Characterization of Blood and Pancreatic Ductal Adenocarcinoma-Derived Extracellular Vesicles. Methods Mol Biol 2025; 2908:225-238. [PMID: 40304913 DOI: 10.1007/978-1-0716-4434-8_15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2025]
Abstract
Extracellular vesicles (EVs) are phospholipid bilayer-enclosed nanoparticles secreted by most cells into the biofluids. They have an important role in intercellular communication by carrying bioactive components (e.g., lipids, proteins, nucleic acids, and other metabolites) and reflecting the biochemical and metabolic processes of their parent cell. Due to their widespread molecular cargo, they have potential diagnostic and therapeutic roles, so they can be considered as new generation biomarkers. With the drastic increase in the number of publications and the comparability of the obtained results, the development of standardized protocols has become necessary. Here we aim to demonstrate the applicability of two, non-conventional techniques, such as ATR-infrared spectroscopy (ATR-FTIR) and electromagnetic piezoelectric acoustic sensor (EMPAS) in the characterization of compositional changes of blood- and pancreatic tumor-derived vesicles. In addition, we provide guidelines in sample collection, isolation, and separation of EVs. IR spectroscopy, as a fast and label-free technique, gives biochemical insight into the sample composition, while EMPAS serves information on vesicle quantity and in membrane integrity changes during storage.
Collapse
Affiliation(s)
- Imola Cs Szigyártó
- HUN-REN Research Centre for Natural Sciences, Institute of Materials and Environmental Chemistry, Biomolecular Self-assembly Research Group, Budapest, Hungary.
| | - Priyanka Singh
- HUN-REN Research Centre for Natural Sciences, Institute of Materials and Environmental Chemistry, Biomolecular Self-assembly Research Group, Budapest, Hungary
- Hevesy György PhD School of Chemistry, Eötvös Loránd University, Budapest, Hungary
| | - Tasvilla Sonallya
- HUN-REN Research Centre for Natural Sciences, Institute of Materials and Environmental Chemistry, Biomolecular Self-assembly Research Group, Budapest, Hungary
- Hevesy György PhD School of Chemistry, Eötvös Loránd University, Budapest, Hungary
| | - Loránd Románszki
- HUN-REN Research Centre for Natural Sciences, Institute of Materials and Environmental Chemistry, Functional Interfaces Research Group, Budapest, Hungary
| | - Judith Mihály
- HUN-REN Research Centre for Natural Sciences, Institute of Materials and Environmental Chemistry, Biological Nanochemistry Research Group, Budapest, Hungary
- Department of Chemistry, Eszterházy Károly Catholic University, Eger, Hungary
| | - Zoltán Wiener
- Department of Genetics, Cell and Immunobiology, Semmelweis University, Budapest, Hungary
| | - Tímea Bebesi
- Hevesy György PhD School of Chemistry, Eötvös Loránd University, Budapest, Hungary
- HUN-REN Research Centre for Natural Sciences, Institute of Materials and Environmental Chemistry, Biological Nanochemistry Research Group, Budapest, Hungary
| | - Gábor Mészáros
- HUN-REN Research Centre for Natural Sciences, Institute of Materials and Environmental Chemistry, Functional Interfaces Research Group, Budapest, Hungary
| | - Zsófia Keresztes
- HUN-REN Research Centre for Natural Sciences, Institute of Materials and Environmental Chemistry, Functional Interfaces Research Group, Budapest, Hungary
| | - Michael Thompson
- Department of Chemistry, University of Toronto, Toronto, ON, Canada
| | - Zoltán Varga
- HUN-REN Research Centre for Natural Sciences, Institute of Materials and Environmental Chemistry, Biological Nanochemistry Research Group, Budapest, Hungary
- Depatment of Physical Chemistry and Materials Science, Budapest University of Technology and Economics, Budapest, Hungary
| | - Tamás Beke-Somfai
- HUN-REN Research Centre for Natural Sciences, Institute of Materials and Environmental Chemistry, Biomolecular Self-assembly Research Group, Budapest, Hungary.
| |
Collapse
|
38
|
Liu W, Wang X, Chen Y, Yuan J, Zhang H, Jin X, Jiang Y, Cao J, Wang Z, Yang S, Wang B, Wu T, Li J. Distinct molecular properties and functions of small EV subpopulations isolated from human umbilical cord MSCs using tangential flow filtration combined with size exclusion chromatography. J Extracell Vesicles 2025; 14:e70029. [PMID: 39783889 PMCID: PMC11714183 DOI: 10.1002/jev2.70029] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 12/03/2024] [Accepted: 12/07/2024] [Indexed: 01/12/2025] Open
Abstract
As functional derivatives of mesenchymal stem cells (MSCs), small extracellular vesicles (sEVs) have garnered significant attention and application in regenerative medicine. However, the technical limitations for large-scale isolation of sEVs and their heterogeneous nature have added complexity to their applications. It remains unclear if the heterogeneous sEVs represent different aspects of MSCs functions. Here, we provide a method for the large-scale production of sEVs subpopulations derived from human umbilical cord mesenchymal stem cells (HucMSCs), utilizing tangential flow filtration combined with size exclusion chromatography. The resulting subpopulations, S1-sEVs and S2-sEVs, exhibited stable variations in size, membrane-marked proteins, and carrying cargos, thereby displaying distinct functions both in vitro and in animal disease models. S1-sEVs, that highly expressed CD9, HRS and GPC1, demonstrated a greater immunomodulatory impact, while S2-sEVs with enriched expression of CD63 and FLOT1/2 possessed enhanced capacities in promoting cell proliferation and angiogenesis. These discrepancies are attributed to the specific proteins and miRNAs they contain. Further investigation revealed that the two distinct sEVs subpopulations corresponded to different biological processes: the ESCRT pathway (S1-sEVs) and the ESCRT-independent pathway represented by lipid rafts (S2-sEVs). Therefore, we propose the potential for large-scale isolation and purification of sEVs subpopulations from HucMSCs with distinct functions. This approach may provide advantages for targeted therapeutic interventions in various MSC indications.
Collapse
Affiliation(s)
- Wei Liu
- State Key Laboratory of Reproductive Medicine and offspring healthNanjing Medical UniversityNanjingChina
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Xinyu Wang
- State Key Laboratory of Reproductive Medicine and offspring healthNanjing Medical UniversityNanjingChina
| | - Yating Chen
- State Key Laboratory of Reproductive Medicine and offspring healthNanjing Medical UniversityNanjingChina
| | - Jiapei Yuan
- State Key Laboratory of Reproductive Medicine and offspring healthNanjing Medical UniversityNanjingChina
| | - Huiyu Zhang
- State Key Laboratory of Reproductive Medicine and offspring healthNanjing Medical UniversityNanjingChina
| | - Xin Jin
- Wuxi Maternity and Child Health Care HospitalAffiliated Women's Hospital of Jiangnan UniversityWuxiChina
| | - Yuying Jiang
- State Key Laboratory of Reproductive Medicine and offspring healthNanjing Medical UniversityNanjingChina
| | - Junjing Cao
- State Key Laboratory of Reproductive Medicine and offspring healthNanjing Medical UniversityNanjingChina
| | - Zibin Wang
- Center for Analysis and TestingNanjing Medical UniversityNanjingChina
| | - Shuo Yang
- State Key Laboratory of Reproductive Medicine and offspring healthNanjing Medical UniversityNanjingChina
- Department of Immunology, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical CenterNanjing Medical UniversityNanjingChina
| | - Bingwei Wang
- Department of PharmacologyNanjing University of Chinese MedicineNanjingChina
| | - Tinghe Wu
- Kornelis Bio‐pharmaceutical Company LimitedNanjingChina
| | - Jing Li
- State Key Laboratory of Reproductive Medicine and offspring healthNanjing Medical UniversityNanjingChina
- Innovation Center of Suzhou Nanjing Medical UniversitySuzhouChina
| |
Collapse
|
39
|
Zha Z, Jia C, Zhou R, Yin Q, Hu Y, Huang Z, Peng L, Zhang Y, Qiu X, Chen Y, Zhong Y, Wang Y, Pang M, Lu S, Sheng C, Huang L. Clostridium difficile-derived membrane vesicles promote fetal growth restriction via inhibiting trophoblast motility through PPARγ/RXRα/ANGPTL4 axis. NPJ Biofilms Microbiomes 2024; 10:158. [PMID: 39741137 DOI: 10.1038/s41522-024-00630-5] [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: 07/30/2024] [Accepted: 12/07/2024] [Indexed: 01/02/2025] Open
Abstract
Fetal growth restriction (FGR) is a common complication of pregnancy, which seriously endangers fetal health and still lacks effective therapeutic targets. Clostridium difficile (C. difficile) is associated with fetal birth weight, and its membrane vesicles (MVs) are pathogenic vectors. However, the role of C. difficile and its MVs in FGR remains unclear. Here we found that supplementation with C. difficile altered the characteristics of gut microbiota and reduced the birth weight in mice. Interestingly, C. difficile MVs entered placenta, inhibited trophoblast motility, and induced fetal weight loss in mice. Mechanistically, C. difficile MVs activated the PPAR pathway via enhancing the transcriptional activity of PPARγ promoter, consequently inhibiting trophoblast motility. Moreover, PPARγ expression was significantly elevated in FGR placenta, and negatively correlated with fetal birth weight. Together, our findings reveal the significance of C. difficile and its MVs in FGR, providing new insights into the mechanisms of FGR development.
Collapse
Affiliation(s)
- Zhiqiang Zha
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Chunhong Jia
- Department of Neonatology, Guangzhou Key Laboratory of Neonatal Intestinal Diseases, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Ruisi Zhou
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Qinlan Yin
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yu Hu
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Zhipeng Huang
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Linyu Peng
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yichi Zhang
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Xiaowei Qiu
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Ying Chen
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yawen Zhong
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yu Wang
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Menglan Pang
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Shijing Lu
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Chao Sheng
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China.
| | - Liping Huang
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
40
|
Chen W, Xie Y, Xu Z, Shang Y, Yang W, Wang P, Wu Z, Cai G, Hong L. Identification and Functional Analysis of miRNAs in Extracellular Vesicles of Semen Plasma from High- and Low-Fertility Boars. Animals (Basel) 2024; 15:40. [PMID: 39794983 PMCID: PMC11718777 DOI: 10.3390/ani15010040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 12/19/2024] [Accepted: 12/24/2024] [Indexed: 01/13/2025] Open
Abstract
Artificial insemination (AI), as an efficient assisted reproduction technology, can help the livestock industry to improve livestock and poultry breeds, optimize production performance and improve reproductive efficiency. AI technology has been widely used in pig production in China, but boar fertility affects the effectiveness of AI, and more and more studies have shown that there are significant differences in the fertility of boars with similar semen quality indicators. Therefore, this study aimed to identify biomarker molecules that indicate the level of boar fertility, which is important for improving the efficiency of AI. In this study, we collected 40 mL of ejaculates per boar used for extracellular vesicle (EV) characterization in 20 boars and identified 53 differentially expressed miRNAs by small RNA sequencing, of which 44 miRNAs were up-regulated in the high-fertility seminal EVs compared with low-fertility seminal EVs, and nine miRNAs were down-regulated. miR-26a was most significantly down-regulated in the high-fertility group compared to the low-fertility group, and it was hypothesized that this miRNA could be used as a biomolecular marker of semen reproductive performance. To further determine the effect of miR-26a on sperm function, we successfully established a miR-26a overexpression model and found that miR-26a reduced sperm viability, motility, acrosome integrity, plasma membrane integrity and ATP levels. Bioinformatics analysis and dual luciferase reporter analysis revealed that miR-26a directly targets High mobility group A1 (HMGA1). In conclusion, miR-26a can be used as a biomarker to identify high and low fertility in boar semen.
Collapse
Affiliation(s)
- Weidong Chen
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (W.C.); (Y.X.); (Y.S.); (W.Y.); (P.W.); (Z.W.)
| | - Yanshe Xie
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (W.C.); (Y.X.); (Y.S.); (W.Y.); (P.W.); (Z.W.)
| | - Zhiqian Xu
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471023, China;
| | - Yijun Shang
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (W.C.); (Y.X.); (Y.S.); (W.Y.); (P.W.); (Z.W.)
| | - Wenzheng Yang
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (W.C.); (Y.X.); (Y.S.); (W.Y.); (P.W.); (Z.W.)
| | - Pengyao Wang
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (W.C.); (Y.X.); (Y.S.); (W.Y.); (P.W.); (Z.W.)
| | - Zhenfang Wu
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (W.C.); (Y.X.); (Y.S.); (W.Y.); (P.W.); (Z.W.)
- Yunfu Subcenter of Guangdong Laboratory for Lingnan Modern Agriculture, Yunfu 527300, China
- Key Laboratory of South China Modern Biological Seed Industry, Ministry of Agriculture and Rural Affairs, Guangzhou 510520, China
| | - Gengyuan Cai
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (W.C.); (Y.X.); (Y.S.); (W.Y.); (P.W.); (Z.W.)
- Yunfu Subcenter of Guangdong Laboratory for Lingnan Modern Agriculture, Yunfu 527300, China
- Key Laboratory of South China Modern Biological Seed Industry, Ministry of Agriculture and Rural Affairs, Guangzhou 510520, China
- National Regional Gene Bank of Livestock and Poultry (Gene Bank of Guangdong Livestock and Poultry), Guangzhou 510642, China
| | - Linjun Hong
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (W.C.); (Y.X.); (Y.S.); (W.Y.); (P.W.); (Z.W.)
- Key Laboratory of South China Modern Biological Seed Industry, Ministry of Agriculture and Rural Affairs, Guangzhou 510520, China
- National Regional Gene Bank of Livestock and Poultry (Gene Bank of Guangdong Livestock and Poultry), Guangzhou 510642, China
| |
Collapse
|
41
|
Sun Y, He X, Han J, Yin W, Wang H, Li J, Liu W, Kuai X, Lv J, Ji J. Activated hepatic stellate cell-derived small extracellular vesicles facilitate M2 macrophage polarization and hepatoma progression via miR-27a-3p. Front Immunol 2024; 15:1489679. [PMID: 39742261 PMCID: PMC11685157 DOI: 10.3389/fimmu.2024.1489679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Accepted: 12/03/2024] [Indexed: 01/03/2025] Open
Abstract
The progression of hepatoma is heavily influenced by the microenvironment. Tumor-associated macrophages (TAMs) are considered to play a critical role in the tumor microenvironment (TME) and increase the aggressiveness of hepatoma. The activation of hepatic stellate cells (HSCs) is involved in hepatoma progression, and accumulating evidence demonstrates a change in microRNA (miRNA) expression during HSC activation. Therefore, the potential roles of HSCs-related miRNAs in macrophage differentiation and hepatoma progression deserve to be explored. The present study aimed to investigate the effects of miRNAs carried by small extracellular vesicles (sEVs) released by activated HSCs on hepatoma progression. The results indicated that miR-27a-3p was significantly upregulated in cells and corresponding sEVs during the activation of primary rat HSCs and human HSC line-LX2 cells. Furthermore, miR-27a-3p contributed to the proliferation and migration of hepatoma cells and promoted M2 polarization of macrophage. HSC-sEVs overexpressing miR-27a-3p can directly facilitate tumor progression and modulate macrophage polarization, indirectly contributing to hepatoma progression. Finally, Sprouty2 (SPRY2) was verified to be the target gene of miR-27a-3p. In conclusion, activated HSC-derived sEVs with high levels of miR-27a-3p might induce M2 macrophage polarization and promote hepatoma progression, providing new insights into the mechanism of hepatoma progression.
Collapse
Affiliation(s)
- Yufeng Sun
- Department of Pathology, Medical School of Nantong University, Nantong, China
- Key Laboratory of Microenvironment and Translational Cancer Research, Nantong, China
| | - Xiaoqian He
- Department of Pathology, Medical School of Nantong University, Nantong, China
- Key Laboratory of Microenvironment and Translational Cancer Research, Nantong, China
| | - Jiayi Han
- Department of Pathology, Medical School of Nantong University, Nantong, China
- Key Laboratory of Microenvironment and Translational Cancer Research, Nantong, China
| | - Wenxuan Yin
- Department of Pathology, Medical School of Nantong University, Nantong, China
| | - Haichen Wang
- Department of Pathology, Medical School of Nantong University, Nantong, China
| | - Jing Li
- Department of Pathology, Medical School of Nantong University, Nantong, China
| | - Weiqi Liu
- Department of Pathology, Medical School of Nantong University, Nantong, China
| | - Xingwang Kuai
- Department of Pathology, Medical School of Nantong University, Nantong, China
- Key Laboratory of Microenvironment and Translational Cancer Research, Nantong, China
| | - Jiaying Lv
- Department of Pathology, Medical School of Nantong University, Nantong, China
- Key Laboratory of Microenvironment and Translational Cancer Research, Nantong, China
| | - Juling Ji
- Department of Pathology, Medical School of Nantong University, Nantong, China
- Key Laboratory of Microenvironment and Translational Cancer Research, Nantong, China
| |
Collapse
|
42
|
Liu Z, Pang B, Wang Y, Zheng J, Li Y, Jiang J. Advances of New Extracellular Vesicle Isolation and Detection Technologies in Cancer Diagnosis. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024:e2405872. [PMID: 39676429 DOI: 10.1002/smll.202405872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Revised: 11/25/2024] [Indexed: 12/17/2024]
Abstract
Cancer is a global health issue threatening people's lives. Currently, cancer detection methods still have a lot of room for improvement in both efficiency and accuracy. The development and application of new technologies are urgently required for early cancer diagnosis and prognosis. Extracellular vesicles (EVs) are a type of phospholipid bilayer vesicle secreted by cells and play an important role in cancer development and metastasis. These small vesicles participate in cancer information transmission, antigen presentation, angiogenesis, immune response, tumor invasion, and mediate signaling pathways in the tumor microenvironment. Liquid biopsy of EV cargo contents is a fast-developing research area, holding promise for early cancer diagnosis and monitoring cancer progression in real-time. However, current EV detection technologies for clinical translation are still facing many challenges. Recent advancements in developing techniques for EV isolation and detection have made significant progress and are paving the way toward clinical application. Here, the advantages and limitations of traditional EV detection and isolation technologies in cancer diagnosis and prognosis are reviewed. The review also focuses on emerging EV detection and isolation technologies in cancer, discusses the challenges faced by current methods, and explores the perspective of new EV detection techniques for future cancer diagnosis.
Collapse
Affiliation(s)
- Zhihan Liu
- The First Affiliated Hospital of Ningbo University, Health Science Center, Ningbo University, Ningbo, Zhejiang, 315211, China
- Ningbo Clinical Research Center for Urological Disease, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, 315010, China
- Translational Research Laboratory for Urology, Department of Urology, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, 315010, China
| | - Bairen Pang
- The First Affiliated Hospital of Ningbo University, Health Science Center, Ningbo University, Ningbo, Zhejiang, 315211, China
- Ningbo Clinical Research Center for Urological Disease, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, 315010, China
- Translational Research Laboratory for Urology, Department of Urology, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, 315010, China
- Zhejiang Engineering Research Center of Innovative Technologies and Diagnostic and Therapeutic Equipment for Urinary System Diseases, Ningbo, Zhejiang, 315010, China
| | - Yuhui Wang
- Laboratory of Advanced Theranostic Materials and Technology, Ningbo Institute of Materials Technology and Engineering (NIMTE) of the Chinese, Chinese Academy of Sciences, Ningbo, 315000, China
| | - Jianping Zheng
- Laboratory of Advanced Theranostic Materials and Technology, Ningbo Institute of Materials Technology and Engineering (NIMTE) of the Chinese, Chinese Academy of Sciences, Ningbo, 315000, China
| | - Yong Li
- Cancer Care Centre, St. George Hospital, Kogarah, NSW, 2217, Australia
- St. George and Sutherland Clinical Campuses, School of Clinical Medicine UNSW Sydney, Kensington, NSW, 2052, Australia
| | - Junhui Jiang
- The First Affiliated Hospital of Ningbo University, Health Science Center, Ningbo University, Ningbo, Zhejiang, 315211, China
- Ningbo Clinical Research Center for Urological Disease, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, 315010, China
- Translational Research Laboratory for Urology, Department of Urology, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, 315010, China
- Zhejiang Engineering Research Center of Innovative Technologies and Diagnostic and Therapeutic Equipment for Urinary System Diseases, Ningbo, Zhejiang, 315010, China
| |
Collapse
|
43
|
Isogai T, Hirosawa KM, Suzuki KGN. Recent Advancements in Imaging Techniques for Individual Extracellular Vesicles. Molecules 2024; 29:5828. [PMID: 39769916 PMCID: PMC11728280 DOI: 10.3390/molecules29245828] [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: 11/14/2024] [Revised: 12/04/2024] [Accepted: 12/09/2024] [Indexed: 01/16/2025] Open
Abstract
Extracellular vesicles (EVs), secreted from most cells, are small lipid membranes of vesicles of 30 to 1000 nm in diameter and contain nucleic acids, proteins, and intracellular organelles originating from donor cells. EVs play pivotal roles in intercellular communication, particularly in forming niches for cancer cell metastasis. However, EVs derived from donor cells exhibit significant heterogeneity, complicating the investigation of EV subtypes using ensemble averaging methods. In this context, we highlight recent studies that characterize individual EVs using advanced techniques, including single-fluorescent-particle tracking, single-metal-nanoparticle tracking, single-non-label-particle tracking, super-resolution microscopy, and atomic force microscopy. These techniques have facilitated high-throughput analyses of the properties of individual EV particles such as their sizes, compositions, and physical properties. Finally, we address the challenges that need to be resolved via single-particle (-molecule) imaging and super-resolution microscopy in future research.
Collapse
Affiliation(s)
- Tatsuki Isogai
- The United Graduate School of Agricultural Science, Gifu University, Gifu 501-1193, Japan;
| | - Koichiro M. Hirosawa
- Institute for Glyco-Core Research (iGCORE), Gifu University, Gifu 501-1193, Japan;
| | - Kenichi G. N. Suzuki
- The United Graduate School of Agricultural Science, Gifu University, Gifu 501-1193, Japan;
- Institute for Glyco-Core Research (iGCORE), Gifu University, Gifu 501-1193, Japan;
- Division of Advanced Bioimaging, National Cancer Center Research Institute (NCCRI), Tokyo 104-0045, Japan
| |
Collapse
|
44
|
Steć A, Heinz A, Dziomba S. Characterization of extracellular vesicles by capillary zone electrophoresis: A novel concept for characterization of a next-generation drug delivery platform. J Pharm Anal 2024; 14:101004. [PMID: 39802401 PMCID: PMC11721263 DOI: 10.1016/j.jpha.2024.101004] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 05/14/2024] [Accepted: 05/17/2024] [Indexed: 01/16/2025] Open
Abstract
Extracellular vesicles (EVs) are a part of a cell-to-cell communication system of prokaryotic and eukaryotic organisms. Their ability to penetrate biological barriers and to transfer molecules between cells shows their potential as a novel class of drug delivery platform. However, because of the great heterogeneity of EVs and the complexity of biological matrices from which they are typically isolated, reliable quality control procedures need to be established to ensure their safety for medical use. According to current recommendations, quantification of EVs, confirmation of their identity, and purity assessment require the use of several analytical techniques, including particle-size distribution analysis, proteomics, and electron microscopy, making the characterization process demanding. Capillary electrophoresis (CE) has recently emerged as an alternative tool for EV characterization. In this study, the available literature on this novel concept for EV characterization was reviewed. Its performance was critically evaluated and compared with currently used methods. The utility of CE in the quality control of EV-based medicines was discussed.
Collapse
Affiliation(s)
- Aleksandra Steć
- Department of Toxicology, Faculty of Pharmacy, Medical University of Gdansk, 80-416 Gdansk, Poland
| | - Andrea Heinz
- LEO Foundation Center for Cutaneous Drug Delivery, Department of Pharmacy, University of Copenhagen, 2100 Copenhagen, Denmark
| | - Szymon Dziomba
- Department of Toxicology, Faculty of Pharmacy, Medical University of Gdansk, 80-416 Gdansk, Poland
| |
Collapse
|
45
|
Zou Y, Gao B, Lu J, Zhang K, Zhai M, Yuan Z, Aschner M, Chen J, Luo W, Wang L, Zhang J. Long non-coding RNA CASC15 enhances learning and memory in mice by promoting synaptic plasticity in hippocampal neurons. EXPLORATION (BEIJING, CHINA) 2024; 4:20230154. [PMID: 39713210 PMCID: PMC11655312 DOI: 10.1002/exp.20230154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Accepted: 02/01/2024] [Indexed: 12/24/2024]
Abstract
Alzheimer's disease (AD) is a debilitating systemic disorder that has a detrimental impact on the overall well-being of individuals. Emerging research suggests that long non-coding RNAs play a role in neural development and function. Nevertheless, the precise relationship between lncRNAs and Alzheimer's disease remains uncertain. The authors' recent discoveries have uncovered an unconventional mechanism involving the regulation of synaptic plasticity and the functioning of the hippocampal fragile X mental retardation protein 1 (FMR1)-neurotrophin 3 (NTF3) pathway, which is mediated by cancer susceptibility candidate 15 (CASC15). Subsequently, functional rescue experiments were performed to illustrate the efficient delivery of exosomes harboring a significant amount of 2610307p16Rik transcripts, which is the murine equivalent of human CASC15, to the hippocampal region of mice. This resulted in significant improvements in synaptic morphological plasticity and cognitive function in APP/PS1 mice. Given the pivotal involvement of CASC15 in synaptic plasticity and the distinctive regulatory mechanisms of the CASC15-FMR1-NTF3 axis, CASC15 emerges as a promising biomarker for Alzheimer's disease and may even possess potential as a feasible therapeutic target.
Collapse
Affiliation(s)
- Yuankang Zou
- Department of Occupational and Environmental Health and the Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public HealthFourth Military Medical UniversityXi'anChina
| | - Bo Gao
- Institute of Orthopaedic SurgeryXijing HospitalFourth Military Medical UniversityXi'anChina
| | - Jiaqiao Lu
- Department of Occupational and Environmental Health and the Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public HealthFourth Military Medical UniversityXi'anChina
| | - Keying Zhang
- Department of UrologyXijing HospitalFourth Military Medical UniversityXi'anChina
| | - Maodeng Zhai
- Department of Occupational and Environmental Health and the Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public HealthFourth Military Medical UniversityXi'anChina
| | - Ziyan Yuan
- Institute of Medical Information and LibraryChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Michael Aschner
- Department of Molecular PharmacologyAlbert Einstein College of MedicineBronxNew YorkUSA
| | - Jingyuan Chen
- Department of Occupational and Environmental Health and the Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public HealthFourth Military Medical UniversityXi'anChina
| | - Wenjing Luo
- Department of Occupational and Environmental Health and the Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public HealthFourth Military Medical UniversityXi'anChina
| | - Lei Wang
- Department of Medical Research Center, Clinical Medical CollegeYangzhou UniversityYangzhouChina
| | - Jianbin Zhang
- Department of Occupational and Environmental Health and the Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public HealthFourth Military Medical UniversityXi'anChina
| |
Collapse
|
46
|
Thieron H, Krassini L, Kwon S, Fricke S, Nasfi S, Oberkofler L, Ruf A, Kehr J, Kogel K, Weiberg A, Feldbrügge M, Robatzek S, Panstruga R. Practical advice for extracellular vesicle isolation in plant-microbe interactions: Concerns, considerations, and conclusions. J Extracell Vesicles 2024; 13:e70022. [PMID: 39665314 PMCID: PMC11635479 DOI: 10.1002/jev2.70022] [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: 06/12/2024] [Accepted: 11/24/2024] [Indexed: 12/13/2024] Open
Abstract
In recent years, extracellular vesicles (EVs) have emerged as novel key players in plant-microbe interactions. While it is immensely useful to draw on the established "minimal information for studies of extracellular vesicles" (MISEV) guidelines and precedents in mammalian systems, working with plants and their associated microbes poses specific challenges. To navigate researchers through these obstacles, we offer detailed step-by-step suggestions for those embarking on EV research in the context of plant-microbe interactions. The advice is based on recent publications and our collective experience from the diverse plant and microbe systems studied in a dedicated research consortium. We provide considerations for experimental design, optimization, quality control, and recommendations on how to increase yield, purity, and reproducibility of EV isolation. With this perspective article, we aim not only to assist researchers in our field but also to promote discussions on plant and microbe EVs in the broader EV community.
Collapse
Affiliation(s)
- Hannah Thieron
- Unit for Plant Molecular Cell Biology, Institute for Biology IRWTH Aachen UniversityAachenGermany
| | - Laura Krassini
- LMU Munich BiocenterLudwig‐Maximilian‐University of MunichMunichGermany
| | - Seomun Kwon
- Institute for Microbiology, Cluster of Excellence on Plant SciencesHeinrich‐Heine University DüsseldorfDüsseldorfGermany
| | - Sebastian Fricke
- Institute of Plant Science and Microbiology, Department of BiologyUniversität HamburgHamburgGermany
| | - Sabrine Nasfi
- Institute of Phytopathology, Research Centre for BioSystemsLand Use and Nutrition, Justus‐Liebig‐University GiessenGiessenGermany
| | - Lorenz Oberkofler
- LMU Munich BiocenterLudwig‐Maximilian‐University of MunichMunichGermany
| | - Alessa Ruf
- LMU Munich BiocenterLudwig‐Maximilian‐University of MunichMunichGermany
| | - Julia Kehr
- Institute of Plant Science and Microbiology, Department of BiologyUniversität HamburgHamburgGermany
| | - Karl‐Heinz Kogel
- Institute of Phytopathology, Research Centre for BioSystemsLand Use and Nutrition, Justus‐Liebig‐University GiessenGiessenGermany
| | - Arne Weiberg
- LMU Munich BiocenterLudwig‐Maximilian‐University of MunichMunichGermany
| | - Michael Feldbrügge
- Institute for Microbiology, Cluster of Excellence on Plant SciencesHeinrich‐Heine University DüsseldorfDüsseldorfGermany
| | - Silke Robatzek
- LMU Munich BiocenterLudwig‐Maximilian‐University of MunichMunichGermany
| | - Ralph Panstruga
- Unit for Plant Molecular Cell Biology, Institute for Biology IRWTH Aachen UniversityAachenGermany
| |
Collapse
|
47
|
Nguyen KN, Graner AN, Fringuello AR, Zizzo Z, Valenzuela L, Anyanwu K, Lillehei KO, Youssef AS, Guzman S, Coughlan C, Graner MW. Extracellular Vesicles from a Novel Chordoma Cell Line, ARF-8, Promote Tumorigenic Microenvironmental Changes When Incubated with the Parental Cells and with Human Osteoblasts. Int J Mol Sci 2024; 25:12731. [PMID: 39684443 DOI: 10.3390/ijms252312731] [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: 10/25/2024] [Revised: 11/21/2024] [Accepted: 11/21/2024] [Indexed: 12/18/2024] Open
Abstract
Chordomas are rare, generally slow-growing spinal tumors that nonetheless exhibit progressive characteristics over time, leading to malignant phenotypes and high recurrence rates, despite maximal therapeutic interventions. The tumors are notoriously resistant to therapies and are often located in regions that complicate achieving gross total resections. Cell lines from these tumors are rare as well. We cultured a new chordoma cell line (ARF-8) derived from an extensive clival chordoma that extended back to the cervical spine. We characterized the ARF-8 cellular and extracellular vesicle (EV) proteomes, as well as the impacts of ARF-8 EVs on the proteomes and secretomes of recipient cells (both ARF-8 and human osteoblasts) in autocrine and paracrine settings. Our proteomic analyses suggested roles for transforming growth factor beta (TGFB/TGFβ), cell-matrix interactions involving the epithelial-to-mesenchymal transition (EMT), and cell-extracellular matrix interactions in cell migration, consistent with a migratory/metastatic tumor phenotype. We demonstrated that ARF-8 tumor cell migration was dependent on general (arginine-glycine-aspartic acid [RGD]-based) integrin activity and that ARF-8 EVs could promote such migration. ARF-8 EVs also prompted proteomic/secretomic changes in human osteoblast cells, again with indications that cell-cell and cell-extracellular matrix interactions would be activated. All the characteristics typically associated with chordomas as cancers-migration and invasion, therapeutic resistance, metastatic potential-can be driven by tumor EVs. Overall, ARF-8 EVs promoted predicted tumorigenic phenotypes in recipient cells and suggested novel therapeutic targets for chordomas.
Collapse
Affiliation(s)
- Khoa N Nguyen
- Department of Neurosurgery, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA
- School of Medicine, University of Colorado, 13001 E 17th Pl, Aurora, CO 80045, USA
| | - Arin N Graner
- Department of Neurosurgery, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Anthony R Fringuello
- Department of Neurosurgery, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Zoe Zizzo
- Department of Biochemistry, Colorado College, 14 E Cache La Poudre St., Colorado Springs, CO 80903, USA
| | - Lorena Valenzuela
- Department of Biomedical Sciences, Regis University, 3333 Regis Blvd., Denver, CO 80221, USA
| | - Kamara Anyanwu
- Department of Biomedical Sciences, Claremont McKenna College, 888 N Columbia Ave., Claremont, CA 91711, USA
| | - Kevin O Lillehei
- Department of Neurosurgery, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - A Samy Youssef
- Department of Neurosurgery, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Samuel Guzman
- Department of Pathology, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Christina Coughlan
- Department of Neurology, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Michael W Graner
- Department of Neurosurgery, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA
| |
Collapse
|
48
|
Nuzzo D, Girgenti A, Palumbo L, Naselli F, Bavetta M, Marfia G, Picone P. Vesicles: New Advances in the Treatment of Neurodegenerative Diseases. Int J Mol Sci 2024; 25:12672. [PMID: 39684383 DOI: 10.3390/ijms252312672] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 11/16/2024] [Accepted: 11/21/2024] [Indexed: 12/18/2024] Open
Abstract
Neurodegenerative diseases are characterized by brain lesions that limit normal daily activities and represent a major challenge to healthcare systems worldwide, with a significant economic impact. Nanotechnology is the science of manipulating matter at the nanoscale, where materials exhibit unique properties that are significantly different from their larger counterparts. These properties can be exploited for a wide range of applications, including medicine. Among the emerging therapeutic approaches for the treatment of neurodegenerative diseases, nanotechnologies are gaining prominence as a promising avenue to explore. Here, we review the state of the art of biological and artificial vesicles and their biological properties in the context of neurodegenerative diseases. Indeed, nanometric structures such as extracellular vesicles and artificial vesicles represent a promising tool for the treatment of such disorders due to their size, biocompatibility, and ability to transport drugs, proteins, and genetic material across the blood-brain barrier to target specific cells and brain areas. In the future, a deeper and broader synergy between materials science, bioengineering, biology, medicine, and the discovery of new, increasingly powerful delivery systems will certainly enable a more applied use of nanotechnology in the treatment of brain disorders.
Collapse
Affiliation(s)
- Domenico Nuzzo
- Institute for Biomedical Research and Innovation, CNR, Via U. La Malfa 153, 90146 Palermo, Italy
| | - Antonella Girgenti
- Institute for Biomedical Research and Innovation, CNR, Via U. La Malfa 153, 90146 Palermo, Italy
| | - Laura Palumbo
- Institute for Biomedical Research and Innovation, CNR, Via U. La Malfa 153, 90146 Palermo, Italy
| | - Flores Naselli
- Institute for Biomedical Research and Innovation, CNR, Via U. La Malfa 153, 90146 Palermo, Italy
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Viale delle Scienze Building 16, 90128 Palermo, Italy
| | - Martina Bavetta
- Institute for Biomedical Research and Innovation, CNR, Via U. La Malfa 153, 90146 Palermo, Italy
| | - Giovanni Marfia
- Laboratory of Experimental Neurosurgery and Cell Therapy, Neurosurgery Unit, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Pasquale Picone
- Institute for Biomedical Research and Innovation, CNR, Via U. La Malfa 153, 90146 Palermo, Italy
| |
Collapse
|
49
|
Gil-Kulik P, Kluz N, Przywara D, Petniak A, Wasilewska M, Frączek-Chudzik N, Cieśla M. Potential Use of Exosomal Non-Coding MicroRNAs in Leukemia Therapy: A Systematic Review. Cancers (Basel) 2024; 16:3948. [PMID: 39682135 DOI: 10.3390/cancers16233948] [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: 10/06/2024] [Revised: 11/19/2024] [Accepted: 11/23/2024] [Indexed: 12/18/2024] Open
Abstract
Leukemia is a heterogeneous group of hematological malignancies. Despite the enormous progress that has been made in the field of hemato-oncology in recent years, there are still many problems related to, among others, disease recurrence and drug resistance, which is why the search for ideal biomarkers with high clinical utility continues. Research shows that exosomes play a critical role in the biology of leukemia and are associated with the drug resistance, metastasis, and immune status of leukemias. Exosomes with their cargo of non-coding RNAs act as a kind of intermediary in intercellular communication and, at the same time, have the ability to manipulate the cell microenvironment and influence the reaction, proliferative, angiogenic, and migratory properties of cells. Exosomal ncRNAs (in particular, circRNAs and microRNAs) appear to be promising cell-free biomarkers for diagnostic, prognostic, and treatment monitoring of leukemias. This review examines the expression of exosomal ncRNAs in leukemias and their potential regulatory role in leukemia therapy but also in conditions such as disease relapse, drug resistance, metastasis, and immune status. Given the key role of ncRNAs in regulating gene networks and intracellular pathways through their ability to interact with DNA, transcripts, and proteins and identifying their specific target genes, defining potential functions and therapeutic strategies will provide valuable information.
Collapse
Affiliation(s)
- Paulina Gil-Kulik
- Department of Clinical Genetics, Medical University of Lublin, 11 Radziwillowska Str., 20-080 Lublin, Poland
| | - Natalia Kluz
- Department of Clinical Genetics, Medical University of Lublin, 11 Radziwillowska Str., 20-080 Lublin, Poland
- Department of Gastroenterology and Internal Medicine, Medical University of Warsaw, 02-091 Warsaw, Poland
| | - Dominika Przywara
- Department of Clinical Genetics, Medical University of Lublin, 11 Radziwillowska Str., 20-080 Lublin, Poland
| | - Alicja Petniak
- Department of Clinical Genetics, Medical University of Lublin, 11 Radziwillowska Str., 20-080 Lublin, Poland
| | - Małgorzata Wasilewska
- Department of Physical Chemistry, Institute of Chemical Sciences, Faculty of Chemistry, Maria Curie-Sklodowska University in Lublin, Maria Curie-Sklodowska Sq. 3, 20-031 Lublin, Poland
| | - Natalia Frączek-Chudzik
- Institute of Medical Science, College of Medical Science, University of Rzeszow, 35-959 Rzeszow, Poland
| | - Marek Cieśla
- Institute of Medical Science, College of Medical Science, University of Rzeszow, 35-959 Rzeszow, Poland
| |
Collapse
|
50
|
Plavchak CL, Werner AZ, Betz E, Salvachúa D, Beckham GT, Kim Ratanathanawongs Williams S. Determination of particle number concentration for biological particles using AF4-MALS: Dependencies on light scattering model and refractive index. J Chromatogr A 2024; 1737:465460. [PMID: 39476775 DOI: 10.1016/j.chroma.2024.465460] [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: 09/16/2024] [Revised: 10/17/2024] [Accepted: 10/18/2024] [Indexed: 11/10/2024]
Abstract
Determining accurate counts and size distributions for biological particles (bioparticles) is crucial in wide-ranging fields, but current methods to this end are susceptible to bias from polydispersity in size. This bias can be mitigated by incorporating a separation step prior to characterization. For this reason, asymmetrical flow field-flow fractionation (AF4) with on-line multiangle light scattering (MALS) has become an important platform for determining particle size. AF4-MALS has also been increasingly used to report particle concentration, particularly for complex biological particles, yet the impact of light scattering models and particle refractive indices (RI) have not been quantitatively evaluated. Here, we develop an analysis workflow using AF4-MALS to simultaneously separate and determine particles sizes and concentrations. The impacts of the MALS particle counting model used to process data and the chosen RI value(s) on particle counts are systematically assessed for polystyrene latex (PSL) particles and bacterial outer membrane vesicles (OMVs) in the 20-500 nm size range. Across spherical models, PSL and OMV particle counts varied up to 13 % or 200 %, respectively. For the coated-sphere model used in the analysis of OMV samples, the sphere RI value greatly impacts particle counts. As the sphere RI value approaches the RI of the suspending medium, the model becomes increasingly sensitive to the light scattering signal-to-noise ratio ultimately causing erroneous particle counts. Overall, this work establishes the importance of selecting appropriate MALS models and RI values for bioparticles to obtain accurate counts and provides an AF4-MALS method to separate, enumerate, and size polydisperse bioparticles.
Collapse
Affiliation(s)
- Christine L Plavchak
- Department of Chemistry, Colorado School of Mines, 1500 Illinois St., Golden, CO 80401, United States
| | - Allison Z Werner
- Renewable Resources and Enabling Sciences Center, National Renewable Energy Laboratory, Golden, CO 80401, United States
| | - Elizabeth Betz
- Department of Chemistry, Colorado School of Mines, 1500 Illinois St., Golden, CO 80401, United States
| | - Davinia Salvachúa
- Renewable Resources and Enabling Sciences Center, National Renewable Energy Laboratory, Golden, CO 80401, United States
| | - Gregg T Beckham
- Renewable Resources and Enabling Sciences Center, National Renewable Energy Laboratory, Golden, CO 80401, United States
| | | |
Collapse
|