1
|
Chen J, Li Y, Quan X, Chen J, Han Y, Yang L, Zhou M, Mok GSP, Wang R, Zhao Y. Utilizing engineered extracellular vesicles as delivery vectors in the management of ischemic stroke: a special outlook on mitochondrial delivery. Neural Regen Res 2025; 20:2181-2198. [PMID: 39101653 PMCID: PMC11759020 DOI: 10.4103/nrr.nrr-d-24-00243] [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: 02/28/2024] [Revised: 06/03/2024] [Accepted: 06/22/2024] [Indexed: 08/06/2024] Open
Abstract
Ischemic stroke is a secondary cause of mortality worldwide, imposing considerable medical and economic burdens on society. Extracellular vesicles, serving as natural nano-carriers for drug delivery, exhibit excellent biocompatibility in vivo and have significant advantages in the management of ischemic stroke. However, the uncertain distribution and rapid clearance of extracellular vesicles impede their delivery efficiency. By utilizing membrane decoration or by encapsulating therapeutic cargo within extracellular vesicles, their delivery efficacy may be greatly improved. Furthermore, previous studies have indicated that microvesicles, a subset of large-sized extracellular vesicles, can transport mitochondria to neighboring cells, thereby aiding in the restoration of mitochondrial function post-ischemic stroke. Small extracellular vesicles have also demonstrated the capability to transfer mitochondrial components, such as proteins or deoxyribonucleic acid, or their sub-components, for extracellular vesicle-based ischemic stroke therapy. In this review, we undertake a comparative analysis of the isolation techniques employed for extracellular vesicles and present an overview of the current dominant extracellular vesicle modification methodologies. Given the complex facets of treating ischemic stroke, we also delineate various extracellular vesicle modification approaches which are suited to different facets of the treatment process. Moreover, given the burgeoning interest in mitochondrial delivery, we delved into the feasibility and existing research findings on the transportation of mitochondrial fractions or intact mitochondria through small extracellular vesicles and microvesicles to offer a fresh perspective on ischemic stroke therapy.
Collapse
Affiliation(s)
- Jiali Chen
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Taipa, Macao Special Administrative Region, China
| | - Yiyang Li
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Taipa, Macao Special Administrative Region, China
| | - Xingping Quan
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Taipa, Macao Special Administrative Region, China
| | - Jinfen Chen
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Taipa, Macao Special Administrative Region, China
| | - Yan Han
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Taipa, Macao Special Administrative Region, China
| | - Li Yang
- Department of Pharmacy, Hunan Provincial People’s Hospital, the First Affiliated Hospital of Hunan Normal University, Changsha, Hunan Province, China
| | - Manfei Zhou
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Taipa, Macao Special Administrative Region, China
| | - Greta Seng Peng Mok
- Department of Electrical and Computer Engineering, University of Macau, Taipa, Macao Special Administrative Region, China
| | - Ruibing Wang
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Taipa, Macao Special Administrative Region, China
- Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, Taipa, Macao Special Administrative Region, China
| | - Yonghua Zhao
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Taipa, Macao Special Administrative Region, China
- Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, Taipa, Macao Special Administrative Region, China
| |
Collapse
|
2
|
Ji J, Li M, Yan K, Ma J, Wei D, Zhang F, Qiao S, Huang P, Zhang W, Li L, Zheng W, Ren L. circSTIL mediates pirarubicin inhibiting the malignant phenotype of triple-negative breast cancer and acts as a biomarker in plasma exosomes. Mol Immunol 2025; 180:86-95. [PMID: 40022852 DOI: 10.1016/j.molimm.2025.02.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Revised: 02/17/2025] [Accepted: 02/19/2025] [Indexed: 03/04/2025]
Abstract
In clinical practice, pirarubicin (THP) is a widely used triple-negative breast cancer (TNBC) agent. It has been found that circular RNAs (circRNAs) are involved in cancer treatment and progression. However, the biological function of circRNAs in TNBC and the relationship between THP and circRNAs remain poorly studied. circSTIL (hsa_circ_0000069) was screened and validated by bioinformatics analysis, demonstrating that it was highly expressed in TNBC cell lines and plasma exosomes, and correlated with a poor prognosis of patients. The expression level of circSTIL in patients' plasma exosomes has potential diagnostic value in distinguishing TNBC from non-TNBC. In vitro studies confirmed that overexpression of circSTIL promotes the proliferation, migration, and invasion of MDA-MB-231 cells whereas silicification of circSTIL shows the reverse effect. Also, circSTIL mediates THP inhibiting the malignant phenotype of MDA-MB-231 cells. The above results suggested that circSTIL is a possible biomarker for the diagnosis, treatment, and prognosis of TNBC.
Collapse
Affiliation(s)
- Jiahua Ji
- Department of Experimental Pharmacology and Toxicology, School of Pharmaceutical Sciences, Jilin University, 1266 Fujin Road, Changchun, Jilin 130021, China
| | - Min Li
- Department of Experimental Pharmacology and Toxicology, School of Pharmaceutical Sciences, Jilin University, 1266 Fujin Road, Changchun, Jilin 130021, China
| | - Kaixu Yan
- Ultrasound Department, Obstetrics and Gynaecology Hospital of Jilin City, 53, Guanghua Road, Jilin City, Jilin 132000, China
| | - Jiulong Ma
- Department of Experimental Pharmacology and Toxicology, School of Pharmaceutical Sciences, Jilin University, 1266 Fujin Road, Changchun, Jilin 130021, China
| | - Dexian Wei
- Department of Experimental Pharmacology and Toxicology, School of Pharmaceutical Sciences, Jilin University, 1266 Fujin Road, Changchun, Jilin 130021, China
| | - Fan Zhang
- General Surgery Center, Department of Breast Surgery, The First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Sennan Qiao
- Department of Experimental Pharmacology and Toxicology, School of Pharmaceutical Sciences, Jilin University, 1266 Fujin Road, Changchun, Jilin 130021, China
| | - Peng Huang
- School of Agroforestry and Medicine, The Open University of China, Beijing 100000, China
| | - Wenqing Zhang
- Department of Experimental Pharmacology and Toxicology, School of Pharmaceutical Sciences, Jilin University, 1266 Fujin Road, Changchun, Jilin 130021, China
| | - Lu Li
- Department of Experimental Pharmacology and Toxicology, School of Pharmaceutical Sciences, Jilin University, 1266 Fujin Road, Changchun, Jilin 130021, China
| | - Wentao Zheng
- Department of Experimental Pharmacology and Toxicology, School of Pharmaceutical Sciences, Jilin University, 1266 Fujin Road, Changchun, Jilin 130021, China
| | - Liqun Ren
- Department of Experimental Pharmacology and Toxicology, School of Pharmaceutical Sciences, Jilin University, 1266 Fujin Road, Changchun, Jilin 130021, China.
| |
Collapse
|
3
|
Otieno MO, Powrózek T, Garcia-Foncillas J, Martinez-Useros J. The crosstalk within tumor microenvironment and exosomes in pancreatic cancer. Biochim Biophys Acta Rev Cancer 2025; 1880:189308. [PMID: 40180303 DOI: 10.1016/j.bbcan.2025.189308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 03/26/2025] [Accepted: 03/27/2025] [Indexed: 04/05/2025]
Abstract
Pancreatic cancer is one of the most malignant tumors with a grim prognosis. Patients develop chemoresistance that drastically decreases their survival. The chemoresistance is mainly attributed to deficient vascularization of the tumor, intratumoral heterogeneity and pathophysiological barrier due to the highly desmoplastic tumor microenvironment. The interactions of cells that constitute the tumor microenvironment change its architecture into a cancer-permissive environment and stimulate cancer development, metastasis and treatment response. The cell-cell communication in the tumor microenvironment is often mediated by exosomes that harbour a diverse repertoire of molecular cargo, such as proteins, lipids, and nucleic acid, including messenger RNAs, non-coding RNAs and DNA. Therefore, exosomes can serve as potential targets as biomarkers and improve the clinical management of pancreatic cancer to overcome chemoresistance. This review critically elucidates the role of exosomes in cell-cell communication within the tumor microenvironment and how these interactions can orchestrate chemoresistance.
Collapse
Affiliation(s)
- Michael Ochieng' Otieno
- Translational Oncology Division, OncoHealth Institute, Health Research Institute Fundación Jimenez Diaz, Fundación Jimenez Díaz University Hospital, Universidad Autonoma de Madrid (IIS-FJD, UAM), 28040 Madrid, Spain
| | - Tomasz Powrózek
- Department of Human Physiology of the Chair of Preclinical Sciences, Medical University in Lublin, 20-080 Lublin, Poland
| | - Jesus Garcia-Foncillas
- Translational Oncology Division, OncoHealth Institute, Health Research Institute Fundación Jimenez Diaz, Fundación Jimenez Díaz University Hospital, Universidad Autonoma de Madrid (IIS-FJD, UAM), 28040 Madrid, Spain; Medical Oncology Department, Fundación Jimenez Diaz University Hospital, 28040, Madrid, Spain
| | - Javier Martinez-Useros
- Translational Oncology Division, OncoHealth Institute, Health Research Institute Fundación Jimenez Diaz, Fundación Jimenez Díaz University Hospital, Universidad Autonoma de Madrid (IIS-FJD, UAM), 28040 Madrid, Spain; Area of Physiology, Department of Basic Health Sciences, Faculty of Health Sciences, Rey Juan Carlos Univer-Sity, 28922 Madrid, Spain.
| |
Collapse
|
4
|
Tian L, Jin J, Lai F, Yao S, Zhang Y, Liu J, Zhang H, Lu Q, Liu C, Tian S, Lu Y, Liang Y, Zhao Y, Fan H, Ren W. Nebulized M2 macrophage-derived nanovesicles for the treatment of explosion-induced acute lung injury. J Colloid Interface Sci 2025; 691:137381. [PMID: 40187079 DOI: 10.1016/j.jcis.2025.137381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 03/18/2025] [Accepted: 03/18/2025] [Indexed: 04/07/2025]
Abstract
Gas explosion-induced acute lung injury (ALI) presents a significant clinical challenge in industrial and military contexts, yet current therapeutic interventions remain suboptimal. Extracellular vesicles (EVs) have emerged as promising nanoscale therapeutic agents, owing to their superior biocompatibility and inherent therapeutic potential. In this study, we engineered and isolated alternatively activated macrophages extracellular vesicles (M2-EVs) and administered them via nebulization to the injured lung tissue in an established murine model of blast-induced ALI. Administration of M2-EVs led to a significant amelioration of pulmonary function, characterized by decreased lung injury scores and attenuated inflammatory markers. Mechanistically, M2-EVs promoted the M1-to-M2 phenotypic transition of pulmonary macrophages, modulated the P62-Keap1-Nrf2 signaling pathway, and consequently mitigated excessive autophagy and oxidative stress. Collectively, these findings not only offer mechanistic insights into the pathogenesis of blast-induced ALI but also highlight M2-EVs as a promising cell-free therapeutic approach for explosion-related pulmonary injuries.
Collapse
Affiliation(s)
- Linqiang Tian
- Henan Medical Key Laboratory for Research of Trauma and Orthopedics, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan Province 453003, China
| | - Jie Jin
- School of Disaster and Emergency Medicine, Tianjin University, Tianjin 300072, China; Tianjin Key Laboratory of Disaster Medical Technology, Tianjin 300072, China
| | - Feng Lai
- Henan Medical Key Laboratory for Research of Trauma and Orthopedics, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan Province 453003, China
| | - Sanqiao Yao
- Institutes of Health Central Plain, Xinxiang Medical University, Xinxiang, Henan Province 453003, China; School of Public Health, Xinxiang Medical University, Xinxiang, Henan Province 453003, China
| | - Yue Zhang
- Henan Medical Key Laboratory for Research of Trauma and Orthopedics, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan Province 453003, China
| | - Jie Liu
- Henan Medical Key Laboratory for Research of Trauma and Orthopedics, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan Province 453003, China
| | - Huajing Zhang
- School of Disaster and Emergency Medicine, Tianjin University, Tianjin 300072, China; Tianjin Key Laboratory of Disaster Medical Technology, Tianjin 300072, China
| | - Qianying Lu
- School of Disaster and Emergency Medicine, Tianjin University, Tianjin 300072, China; Tianjin Key Laboratory of Disaster Medical Technology, Tianjin 300072, China
| | - Chuanchuan Liu
- School of Disaster and Emergency Medicine, Tianjin University, Tianjin 300072, China; Tianjin Key Laboratory of Disaster Medical Technology, Tianjin 300072, China
| | - Sijia Tian
- School of Disaster and Emergency Medicine, Tianjin University, Tianjin 300072, China; Tianjin Key Laboratory of Disaster Medical Technology, Tianjin 300072, China
| | - Yujia Lu
- School of Disaster and Emergency Medicine, Tianjin University, Tianjin 300072, China; Tianjin Key Laboratory of Disaster Medical Technology, Tianjin 300072, China
| | - Yangfan Liang
- School of Disaster and Emergency Medicine, Tianjin University, Tianjin 300072, China; Tianjin Key Laboratory of Disaster Medical Technology, Tianjin 300072, China
| | - Yanmei Zhao
- School of Disaster and Emergency Medicine, Tianjin University, Tianjin 300072, China; Tianjin Key Laboratory of Disaster Medical Technology, Tianjin 300072, China.
| | - Haojun Fan
- School of Disaster and Emergency Medicine, Tianjin University, Tianjin 300072, China; Tianjin Key Laboratory of Disaster Medical Technology, Tianjin 300072, China.
| | - Wenjie Ren
- Henan Medical Key Laboratory for Research of Trauma and Orthopedics, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan Province 453003, China; Institutes of Health Central Plain, Xinxiang Medical University, Xinxiang, Henan Province 453003, China.
| |
Collapse
|
5
|
Wu F, Li Y, Zhang L, Zhou Y, Xu Y, Cai Y, Ding L, Zhang L, Wang Y, Qian H. Real-Time Isolation and Versatile Detection for Extracellular Vesicles Based on Ordered Porous Layer Interferometry. Anal Chem 2025; 97:5798-5807. [PMID: 40045887 DOI: 10.1021/acs.analchem.4c07108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
Extracellular vesicles (EVs) are progressively becoming novel instruments for clinical therapeutics and liquid biopsies. Due to the complexity of biofluids and the physicochemical properties of EVs, the biological activity, velocity, and efficiency of EV isolation are always unsatisfying. Here, we present a real-time isolation approach of EVs derived from cells and urine using ordered porous layer interferometry with a silica colloidal crystal film as the sensing substrate, achieving efficiency greater than 90%. The online concentration detection function is performed during the isolation process on the basis of its real-time monitoring characteristic. Using membrane protein markers of urine EVs as targets, this technique has a high diagnostic value for liquid biopsy of prostate cancer. Furthermore, we compared multiple EV membrane protein expression and binding dissociation kinetic data from cells and urine. In summary, this multifunctional approach provides a novel strategy for the rapid EVs isolation, concentration detection, drug target screening, and liquid biopsy of various body fluids.
Collapse
Affiliation(s)
- Feng Wu
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, China
| | - Yaoyang Li
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, China
| | - Linlin Zhang
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, China
| | - Yuchen Zhou
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, China
| | - Yili Xu
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, China
| | - Youpeng Cai
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, China
| | - Lingling Ding
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, China
| | - Leilei Zhang
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, China
| | - Yanfeng Wang
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, China
| | - Hui Qian
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, China
| |
Collapse
|
6
|
Salviano-Silva A, Wollmann K, Brenna S, Reimer R, Neumann JE, Dottermusch M, Woythe L, Maire CL, Puig B, Schüller U, Saul MJ, Westphal M, Drexler R, Dührsen L, Gempt J, Heiland DH, Lamszus K, Ricklefs FL. Extracellular Vesicles Carrying Tenascin-C are Clinical Biomarkers and Improve Tumor-Derived DNA Analysis in Glioblastoma Patients. ACS NANO 2025; 19:9844-9859. [PMID: 40056466 PMCID: PMC11924321 DOI: 10.1021/acsnano.4c13599] [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: 03/10/2025]
Abstract
Extracellular vesicles (EVs) act as carriers of biological information from tumors to the bloodstream, enabling the detection of circulating tumor material and tracking of disease progression. This is particularly crucial in glioblastoma, a highly aggressive and heterogeneous tumor that is challenging to monitor. Using imaging flow cytometry (IFCM), we conducted an immunophenotyping analysis of eight glioma-associated antigens and tetraspanins in plasma EVs from 37 newly diagnosed glioblastoma patients (pre- and post-surgery), 11 matched individuals with recurrent glioblastoma, and 22 healthy donors (HD). Tenascin-C (TNC) positive EVs displayed the strongest differences in newly diagnosed and recurrent glioblastoma patients, when compared to non-tumor subjects. Among dual-positive subpopulations, TNC+/CD9+ EVs were the most elevated in newly diagnosed (FC = 7.6, p <0.0001, AUC = 81%) and recurrent patients (FC = 16.5, p <0.0001; AUC = 90%) than HD. In comparison with other CNS tumors (n = 25), this subpopulation was also 34.5-fold higher in glioblastoma than in meningioma cases (p <0.01). Additionally, TNC+/CD9+ EV levels were 3.3-fold elevated in cerebrospinal fluid from glioblastoma patients (n = 6) than controls (p <0.05). Aberrant TNC levels were further observed in glioblastoma EVs from different sources and purified via different methods. Immunohistochemical analysis revealed high levels of TNC in tumor tissues. Spatial transcriptomic analysis indicated a TNC overexpression in malignant cell populations of glioblastoma resections, particularly in cells with mesenchymal-like signatures and chromosomal aberrations. Lastly, we purified TNC+ EVs from plasma of 21 glioblastoma patients by magnetic sorting and detected the oncogenic mutation TERT*C228T by droplet digital PCR. The mutant allele frequency was higher in TNC+ EVs vs TNC-negative EVs (FC = 32, p <0.001), total EVs (FC = 5.3, p <0.001) or cell-free DNA (FC = 5.3, p <0.01). In conclusion, circulating TNC+ EVs may have potential as clinical biomarkers in glioblastoma, and their purification could improve the identification of tumor-specific mutations in liquid biopsies.
Collapse
Affiliation(s)
- Amanda Salviano-Silva
- Department of Neurosurgery, University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany
| | - Kathrin Wollmann
- Department of Neurosurgery, University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany
| | - Santra Brenna
- Neurology Department, Experimental Research in Stroke and Inflammation, University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany
| | - Rudolph Reimer
- Leibniz Institute for Experimental Virology, Hamburg 20251, Germany
| | - Julia E Neumann
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany
- Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany
| | - Matthias Dottermusch
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany
- Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany
| | - Laura Woythe
- Oxford Nanoimaging Limited (ONI), Oxford OX2 8TA, U.K
| | - Cecile L Maire
- Department of Neurosurgery, University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany
| | - Berta Puig
- Neurology Department, Experimental Research in Stroke and Inflammation, University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany
| | - Ulrich Schüller
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany
- Department of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
- Children's Cancer Research Center Hamburg, Hamburg 20246, Germany
| | - Meike J Saul
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, University Cancer Center Hamburg, University Clinic Hamburg-Eppendorf, Hamburg 20246, Germany
| | - Manfred Westphal
- Department of Neurosurgery, University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany
| | - Richard Drexler
- Department of Neurosurgery, University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany
| | - Lasse Dührsen
- Department of Neurosurgery, University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany
| | - Jens Gempt
- Department of Neurosurgery, University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany
| | - Dieter H Heiland
- Department of Neurosurgery, Medical Center University of Freiburg, Freiburg D-79106, Germany
- Translational Neurosurgery, Friedrich-Alexander University Erlangen Nuremberg, Erlangen 91054, Germany
- Department of Neurosurgery, University Hospital Erlangen, Friedrich-Alexander University Erlangen Nuremberg, Erlangen 91054, Germany
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, United States
- German Cancer Consortium (DKTK), Partner Site Freiburg, Freiburg D-79106, Germany
| | - Katrin Lamszus
- Department of Neurosurgery, University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany
| | - Franz L Ricklefs
- Department of Neurosurgery, University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany
| |
Collapse
|
7
|
Chen P, Zeng L, Wang T, He J, Xiong S, Chen G, Wang Q, Chen H, Xie J. The communication role of extracellular vesicles in the osteoarthritis microenvironment. Front Immunol 2025; 16:1549833. [PMID: 40165965 PMCID: PMC11955493 DOI: 10.3389/fimmu.2025.1549833] [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: 12/22/2024] [Accepted: 02/26/2025] [Indexed: 04/02/2025] Open
Abstract
Osteoarthritis (OA) is the most common degenerative joint disease worldwide, characterized by synovial inflammation, cartilage loss, and reactive hyperplasia of subchondral bone, affecting the quality of life of hundreds of millions of people. However, the molecular mechanisms underlying the occurrence and progression of OA remain unclear, and there is no therapy can substantially interrupt or reverse the destructive process of OA. More insight into the pathogenesis of OA may result in innovative therapeutics. The OA microenvironment plays a pivotal role in the development and progression of OA, which encompasses chondrocytes, adipocytes, synovial fibroblasts, endothelial cells, and immune cells. Extracellular vesicles (EVs) have emerged as a novel form of intercellular communication, mediating the transfer of a range of bioactive molecules to create a specific microenvironment. Recent studies have reported that the cargos of EVs play a crucial role in the pathogenesis of OA, including noncoding RNAs (ncRNAs), proteins, and lipids. This review systematically analyzes and summarizes the biological characteristics and functionalities of EVs derived from diverse cellular sources, especially how EVs mediate communication between different cells in the OA microenvironment, with a view to providing new insights into the pathogenesis of OA.
Collapse
Affiliation(s)
- Pu Chen
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Lingfeng Zeng
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
- Guangdong Provincial Key Laboratory of Chinese Medicine for Prevention and Treatment of Refractory Chronic Diseases, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
- Bone and Joint Research Team of Degeneration and Injury, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, China
| | - Ting Wang
- Department of Operating Room, Ji’an County Traditional Chinese Medicine Hospital, Ji’an, Jiangxi, China
| | - Jianbo He
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Shuai Xiong
- School of Anesthesiology, Shandong Second Medical University, Weifang, Shandong, China
| | - Gang Chen
- Department of Orthopedic Surgery, Jiangxi University of Traditional Chinese Medicine Affiliated Hospital, Nanchang, Jiangxi, China
| | - Qingfu Wang
- Department of Orthopedic Surgery, Beijing University of Chinese Medicine Third Affiliated Hospital, Beijing, China
| | - Haiyun Chen
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
- Guangdong Provincial Hospital of Chinese Medicine, Zhuhai Hospital, Zhuhai, China
| | - Jiewei Xie
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| |
Collapse
|
8
|
Yang JJ, He SQ, Huang B, Wang PX, Xu F, Lin X, Liu J. A bibliometric and visualized analysis of extracellular vesicles in degenerative musculoskeletal diseases (from 2006 to 2024). Front Pharmacol 2025; 16:1550208. [PMID: 40183074 PMCID: PMC11966045 DOI: 10.3389/fphar.2025.1550208] [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: 12/23/2024] [Accepted: 02/26/2025] [Indexed: 04/05/2025] Open
Abstract
Background With the rapid development of extracellular vesicles (EVs) in regenerative medicine research, they have become a promising new direction in the mechanistic, diagnosis and treatment studies of degenerative musculoskeletal diseases (DMDs), and has attracted increasing attention. However, there is currently a lack of comprehensive and objective summary analysis to help researchers quickly and conveniently understand the development trajectory and future trends of this field. Method This study collected articles and reviews published from 2006 to 2024 on EVs in DMDs from the Web of Science database. Bibliometric and visual analysis was conducted using several tools, including Microsoft Excel Office, VOSviewer, CiteSpace, Pajek, and R packages. Results 1,182 publications were included in the analysis from 2006 to 2024. Notably, there was a rapid increase in the number of publications starting in 2016, suggesting that this field remains in a developmental stage. Co-authorship analysis revealed that China ranked first in terms of publications, whereas the United States led in citations. The journal with the highest number of publications was International Journal of Molecular Sciences (INT J MOL SCI). The most prolific authors were Ragni, E with 23 publications, while the most cited author was Toh, WS. Additionally, nine of the top 10 institutions were from China, with Shanghai Jiao Tong University leading in the number of publications. The most cited article was "MSC exosomes mediate cartilage repair by enhancing proliferation, attenuating apoptosis and modulating immune reactivity", authored by Zhang, S, and published in BIOMATERIALS in 2018. Conclusion This study, through bibliometric and visual analysis, clearly illustrates the collaborative relationships among countries, authors, institutions, and journals, providing valuable insights for researchers seeking academic collaboration opportunities. Moreover, the analysis of keywords and citations allows researchers to better understand key research hotspots and frontiers in this field, and points toward promising directions for future research. The growing interest in EV research in DMDs over recent years indicates increasing attention and a dynamic progression in this field.
Collapse
Affiliation(s)
- Jun-Jie Yang
- Department of Radiology, The Second Xiangya Hospital, Central South University, Changsha, China
- Department of Radiology, The Second Affiliated Hospital of Xinjiang Medical University, Ürümqi, China
| | - Sha-Qi He
- Department of Radiology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Bei Huang
- Department of Radiology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Peng-Xin Wang
- Department of Radiology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Feng Xu
- National Clinical Research Center for Metabolic Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Xiao Lin
- Department of Radiology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Jun Liu
- Department of Radiology, The Second Xiangya Hospital, Central South University, Changsha, China
- Clinical Research Center for Medical Imaging in Hunan Province, Department of Radiology Quality Control Center in Hunan Province, Changsha, China
| |
Collapse
|
9
|
Wu Q, Wu Q, Lin H, Zhang C, You Z, Kang S, Xu Y, Chen X, Yang C, Song Y, Zhu L. Microfluidic Replication and Phenotypic Profiling of Extracellular Vesicles from the Tumor Microenvironment Using Dual-Switch Aptamer Logic Gates. Anal Chem 2025; 97:5313-5323. [PMID: 40012368 DOI: 10.1021/acs.analchem.5c00234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/28/2025]
Abstract
The phenotypic profiling of extracellular vesicles (EVs) within the tumor microenvironment (TME) provides critical insights into the intercellular communication mechanisms of EVs underlying tumor physiology. However, conventional methods typically isolate EVs from the extracellular space through tissue fragmentation, which compromises tissue viability, and neglects the spatial organization of the tissue and the dynamic nature of EV secretion. Herein, we introduce an innovative microfluidic platform to cultivate intact tumor tissues while preserving their spatial architecture and facilitating natural EV secretion. This system enables the direct replication of EVs onto the chip for high-fidelity phenotypic analysis. Utilizing a combinatorial-aptamer-induced dual-switch logic gate methodology, this approach allows for the precise subtyping of EVs derived from both tumor cells and immune cells within the TME. Specifically, aptamers targeting EpCAM and PD-L1, along with the connector probe, were employed to induce a dual-switch signal to identify distinct EV populations. This strategy enables noninvasive, real-time capture and phenotypic profiling of EVs directly within the microfluidic environment. Furthermore, our findings indicate that immunotherapy with PD-1 antibodies significantly enhances the secretion of EVs by immune cells within the TME, underscoring the potential role of EVs as mediators of therapeutic responses. Overall, we have developed a robust, noninvasive method for the phenotypic profiling of EVs in the TME, offering a powerful tool for investigating the biological functions and implications of EVs in tumor pathophysiology.
Collapse
Affiliation(s)
- Qiaoyi Wu
- Department of Trauma Center and Emergency Surgery, The First Affiliated Hospital of Fujian Medical University, Department of Trauma Center & Emergency Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, P. R. China
| | - Qiuyue Wu
- The MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, P. R. China
| | - Haoting Lin
- The MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, P. R. China
| | - Chi Zhang
- The MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, P. R. China
| | - Zhenlong You
- The MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, P. R. China
| | - Siyin Kang
- The MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, P. R. China
| | - Yuanfeng Xu
- The MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, P. R. China
| | - Xiaofeng Chen
- The MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, P. R. China
- School of Environmental Science and Engineering, Hainan University, Haikou 570228, P. R. China
| | - Chaoyong Yang
- The MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, P. R. China
- Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P. R. China
| | - Yanling Song
- The MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, P. R. China
| | - Lin Zhu
- The MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, P. R. China
- School of Environmental Science and Engineering, Hainan University, Haikou 570228, P. R. China
| |
Collapse
|
10
|
Jin Y, Xu C, Zhu Y, Gu Z. Extracellular vesicle as a next-generation drug delivery platform for rheumatoid arthritis therapy. J Control Release 2025; 381:113610. [PMID: 40058499 DOI: 10.1016/j.jconrel.2025.113610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 03/01/2025] [Accepted: 03/04/2025] [Indexed: 03/16/2025]
Abstract
Rheumatoid arthritis (RA) is a systemic autoimmune disorder characterized by chronic inflammation and progressive damage to connective tissue. It is driven by dysregulated cellular homeostasis, often leading to autoimmune destruction and permanent disability in severe cases. Over the past decade, various drug delivery systems have been developed to enable targeted therapies for disease prevention, reduction, or suppression. As an emerging therapeutic platform, extracellular vesicles (EVs) offer several advantages over conventional drug delivery systems, including biocompatibility and low immunogenicity. Consequently, an increasing number of studies have explored EV-based delivery systems in the treatment of RA, leveraging their natural ability to evade phagocytosis, prolong in vivo half-life, and minimize the immunogenicity of therapeutic agents. In this review, we first provide an in-depth overview of the pathogenesis of RA and the current treatment landscape. We then discuss the classification and biological properties of EVs, their potential therapeutic mechanisms, and the latest advancements in EVs as drug delivery platforms for RA therapy. We emphasize the significance of EVs as carriers in RA treatment and their potential to revolutionize therapeutic strategies. Furthermore, we examine key technological innovations and the future trajectory of EV research, focusing on the challenges and opportunities in translating these platforms into clinical practice. Our discussion aims to offer a comprehensive understanding of the current state and future prospects of EV-based therapeutics in RA.
Collapse
Affiliation(s)
- Yi Jin
- Department of Rheumatology, Research Center of Clinical Medicine, Research Center of Immunology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China
| | - Cong Xu
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, United States
| | - Yujuan Zhu
- Department of Rheumatology, Research Center of Clinical Medicine, Research Center of Immunology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China.
| | - Zhifeng Gu
- Department of Rheumatology, Research Center of Clinical Medicine, Research Center of Immunology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China.
| |
Collapse
|
11
|
Zhang Y, Qin X, Xu Z, Liu W, Lu H, Yang Y, Yang J, Li X, Zhang Y, Yang F. Electric Field-Resistant Bubble-Enhanced Wash-Free Profiling of Extracellular Vesicle Surface Markers. ACS NANO 2025; 19:8093-8107. [PMID: 39985473 DOI: 10.1021/acsnano.4c16353] [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: 02/24/2025]
Abstract
Efficient profiling of circulating extracellular vesicles (EVs) benefits noninvasive cancer diagnosis and therapeutic monitoring, but is technically hampered by tedious isolation, multistep washing, and poor sensitivity. Here, we report multifunctional bubbles that enable self-separation, wash-free, single-step, and ultrasensitive profiling of EV surface markers in plasma samples for early diagnosis and treatment monitoring of lung cancer. In this assay, the buoyancy-dominated bubble is electric field-resistant, allowing EV-responsive release of electroactive probes for electrohydrodynamic nanoshearing force-enhanced hybridization, self-separation from the electrode interface for minimizing noise in electrochemical measurements, and one-step wash-free EV profiling. This assay achieves sensitivity near a single-EV level, shows high specificity against nontarget EVs, and tracks EV phenotypic changes induced by drugs. We further show that this technology can classify plasma samples (n = 111) between cancer patients and noncancer controls with accuracies >95%, enable accurate early diagnosis via machine learning, and monitor pre/post-surgery efficacy with higher accuracy over routine clinical serum markers. This bubble-driven one-step EV assay provides a promising wash-free quantitative tool to enable clinical precision liquid biopsies.
Collapse
Affiliation(s)
- Yuyuan Zhang
- Guangxi Key Laboratory of Pharmaceutical Precision Detection and Screening, Key Laboratory of Micro-Nanoscale Bioanalysis and Drug Screening of Guangxi Education Department, Pharmaceutical College, State Key Laboratory of Targeting Oncology, Guangxi Medical University, Nanning 530021, China
| | - Xiaojie Qin
- Guangxi Key Laboratory of Pharmaceutical Precision Detection and Screening, Key Laboratory of Micro-Nanoscale Bioanalysis and Drug Screening of Guangxi Education Department, Pharmaceutical College, State Key Laboratory of Targeting Oncology, Guangxi Medical University, Nanning 530021, China
| | - Zhanyu Xu
- Department of Thoracic and Cardiovascular Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Weifeng Liu
- Guangxi Key Laboratory of Pharmaceutical Precision Detection and Screening, Key Laboratory of Micro-Nanoscale Bioanalysis and Drug Screening of Guangxi Education Department, Pharmaceutical College, State Key Laboratory of Targeting Oncology, Guangxi Medical University, Nanning 530021, China
| | - Hao Lu
- Guangxi Key Laboratory of Pharmaceutical Precision Detection and Screening, Key Laboratory of Micro-Nanoscale Bioanalysis and Drug Screening of Guangxi Education Department, Pharmaceutical College, State Key Laboratory of Targeting Oncology, Guangxi Medical University, Nanning 530021, China
| | - Yu Yang
- Guangxi Key Laboratory of Pharmaceutical Precision Detection and Screening, Key Laboratory of Micro-Nanoscale Bioanalysis and Drug Screening of Guangxi Education Department, Pharmaceutical College, State Key Laboratory of Targeting Oncology, Guangxi Medical University, Nanning 530021, China
| | - Jie Yang
- Guangxi Key Laboratory of Pharmaceutical Precision Detection and Screening, Key Laboratory of Micro-Nanoscale Bioanalysis and Drug Screening of Guangxi Education Department, Pharmaceutical College, State Key Laboratory of Targeting Oncology, Guangxi Medical University, Nanning 530021, China
| | - Xinchun Li
- Guangxi Key Laboratory of Pharmaceutical Precision Detection and Screening, Key Laboratory of Micro-Nanoscale Bioanalysis and Drug Screening of Guangxi Education Department, Pharmaceutical College, State Key Laboratory of Targeting Oncology, Guangxi Medical University, Nanning 530021, China
| | - Yuanqing Zhang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Fan Yang
- Guangxi Key Laboratory of Pharmaceutical Precision Detection and Screening, Key Laboratory of Micro-Nanoscale Bioanalysis and Drug Screening of Guangxi Education Department, Pharmaceutical College, State Key Laboratory of Targeting Oncology, Guangxi Medical University, Nanning 530021, China
| |
Collapse
|
12
|
Poorkazem H, Saber M, Moradmand A, Yakhkeshi S, Seydi H, Hajizadeh-Saffar E, Shekari F, Hassani SN. Comparative effects of various extracellular vesicle subpopulations derived from clonal mesenchymal stromal cells on cultured fibroblasts in wound healing-related process. Int J Biochem Cell Biol 2025; 180:106737. [PMID: 39828140 DOI: 10.1016/j.biocel.2025.106737] [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: 10/23/2024] [Revised: 12/25/2024] [Accepted: 01/09/2025] [Indexed: 01/22/2025]
Abstract
INTRODUCTION Non-healing wounds pose significant challenges and require effective therapeutic interventions. Extracellular vesicles (EVs) have emerged as promising cell-free therapeutic agents in tissue regeneration. However, the functional differences between different subpopulations of EVs in wound healing remain understudied. This study aimed to evaluate the effects of two distinct subpopulations of clonal mesenchymal stromal cells (cMSC)-derived EVs (cMSC-EVs), namely 20 K and 110K-cMSC-EVs, primarily on in vitro wound healing process, providing fast and cost-effective alternatives to animal models. METHODS In vitro assays were conducted to compare the effects of 20 K and 110K-cMSC-EVs, isolated through high-speed centrifugation and differential ultracentrifugation, respectively. For evaluation the main mechanisms of wound healing, including cell proliferation, cell migration, angiogenesis, and contraction. Human dermal fibroblasts (HDF) were considered as the main cells for analysis of these procedures. Moreover, gene expression analysis was performed to assess the impact of these EV subpopulations on the related process of wound healing on HDF. RESULTS The results demonstrated that both 20 K and 110K-cMSC-EVs exhibited beneficial effects on cell proliferation, cell migration, angiogenesis, and gel contraction. RT-qPCR revealed that both EV types downregulated interleukin 6 (IL6), induced proliferation by upregulating proliferating cell nuclear antigen (PCNA), and regulated remodeling by upregulating matrix metallopeptidase 1 (MMP1) and downregulating collagen type 1 (COL1). DISCUSSION This study highlights the effects of both 20 K and 110K-cMSC-EVs on the potency of HDFs in wound healing-related process. As the notable finding, 20K-cMSC-EVs offer a more feasible and cost-effective subpopulation for isolation and follow the GMP standard, recommended to utilize this fraction for therapeutic application.
Collapse
Affiliation(s)
- Hedie Poorkazem
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran; Department of Developmental Biology, School of Basic Sciences and Advanced Technologies in Biology, University of Science and Culture, Tehran, Iran
| | - Maryam Saber
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Azadeh Moradmand
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Saeed Yakhkeshi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Homeyra Seydi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Ensiyeh Hajizadeh-Saffar
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran; Department of Regenerative Biomedicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Faezeh Shekari
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran; Department of Regenerative Biomedicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| | - Seyedeh-Nafiseh Hassani
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran; Department of Regenerative Biomedicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| |
Collapse
|
13
|
Shakouri-Motlagh A, O'Connor AJ, Brennecke SP, Heath DE, Kalionis B. Extracellular vesicles support increased expansion of mesenchymal stromal cells on fetal membrane-derived decellularized extracellular matrix. Cell Tissue Res 2025; 399:323-336. [PMID: 39715869 DOI: 10.1007/s00441-024-03946-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 12/09/2024] [Indexed: 12/25/2024]
Abstract
Decidual mesenchymal stromal cells (DMSC) were the source of extracellular vesicles (DMSC_EV). The xCELLigence real-time cell growth assay revealed increasing concentrations of EVs decreased DMSC attachment in the early growth phase but stimulated DMSC proliferation at day 7 when grown on tissue culture plastic (TCP). DMSC attachment and proliferation varied depending on the growth surface and DMSC_EV supplementation. DMSC attachment increased on decellularized and solubilized amniotic (s-dAM) whether or not EVs were added. Only Matrigel substrate increased DMSC attachment with added EVs. The addition of EVs increased DMSC proliferation only on the s-dAM substrate. DMSCs were more motile on s-dAM and decellularized and solubilized chorionic (s-dCM) membranes following EV addition. The osteogenic potential of DMSCs was improved on s-dAM substrates when supplanted with EVs. Finally, the levels of reactive oxygen species in DMSCs varied depending on the substrate but not on added EVs. We show that the addition of in vitro EVs isolated from the source being expanded (i.e., DMSCs) and the presence of ECM improve DMSC behaviours during ex vivo expansion. The inclusion of two key components of the MSC niche, EVs and ECM, benefitted the ex vivo expansion of MSCs. Added in vitro EVs increased the proliferation of DMSCs when grown on s-dAM but not on s-dCM, whereas they improved DMSC mobility on both surfaces. Testing different ECMs could be used to promote specific desired characteristics of DMSCs, and different combinations of EVs and ECM may enhance desirable MSC characteristics for specific therapeutic settings.
Collapse
Affiliation(s)
- Aida Shakouri-Motlagh
- Department of Biomedical Engineering, School of Engineering, The University of Melbourne, Parkville, VIC, Australia
- Department of Obstetrics, Gynaecology and Newborn Health, University of Melbourne, Royal Women's Hospital Campus, Parkville, VIC, Australia
| | - Andrea J O'Connor
- Department of Biomedical Engineering, School of Engineering, The University of Melbourne, Parkville, VIC, Australia
| | - Shaun P Brennecke
- Department of Obstetrics, Gynaecology and Newborn Health, University of Melbourne, Royal Women's Hospital Campus, Parkville, VIC, Australia
- Department of Maternal-Fetal Medicine Pregnancy Research Centre, Royal Women's Hospital, Parkville, VIC, Australia
| | - Daniel E Heath
- Department of Biomedical Engineering, School of Engineering, The University of Melbourne, Parkville, VIC, Australia.
| | - Bill Kalionis
- Department of Obstetrics, Gynaecology and Newborn Health, University of Melbourne, Royal Women's Hospital Campus, Parkville, VIC, Australia.
- Department of Maternal-Fetal Medicine Pregnancy Research Centre, Royal Women's Hospital, Parkville, VIC, Australia.
| |
Collapse
|
14
|
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
|
15
|
Tang J, Li D, Wang R, Li S, Xing Y, Yu F. Engineered extracellular vesicles: an emerging nanomedicine therapeutic platform. Chem Commun (Camb) 2025. [PMID: 39969526 DOI: 10.1039/d4cc06501h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/20/2025]
Abstract
The intercellular communication role of extracellular vesicles has been widely proved in various organisms. Compelling evidence has illustrated the involvement of these vesicles in both physiological and pathological processes. Various studies indicate that extracellular vesicles surpass conventional synthetic drug carriers, owing to their abundance in organisms, enhanced targeting ability and low immunogenicity. Therefore, extracellular vesicles have been deemed to be potential drug carriers for the treatment of various diseases, and related studies have increased rapidly. Here, we intend to provide a comprehensive and in-depth review of recent advances in the sources, delivery function, extraction and cargo-loading technologies of extracellular vesicles, as well as their clinical potential in constructing emerging nanomedicine therapeutic platforms. In particular, microfluidic-based isolation and drug-loading technologies, as well as the treatment of various diseases, are highlighted. We also make comparisons between extracellular vesicles and other conventional drug carriers and discuss the challenges in developing drug delivery platforms for clinical translation.
Collapse
Affiliation(s)
- Jingshi Tang
- Key Laboratory of Emergency and Trauma, Ministry of Education, Key Laboratory of Hainan Trauma and Disaster Rescue, Key Laboratory of Haikou Trauma, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou 571199, China.
- Engineering Research Center for Hainan Bio-Smart Materials and Bio-Medical Devices, Key Laboratory of Hainan Functional Materials and Molecular Imaging, College of Pharmacy, College of Emergency and Trauma, Hainan Medical University, Haikou 571199, China
| | - Dezhong Li
- Key Laboratory of Emergency and Trauma, Ministry of Education, Key Laboratory of Hainan Trauma and Disaster Rescue, Key Laboratory of Haikou Trauma, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou 571199, China.
- Engineering Research Center for Hainan Bio-Smart Materials and Bio-Medical Devices, Key Laboratory of Hainan Functional Materials and Molecular Imaging, College of Pharmacy, College of Emergency and Trauma, Hainan Medical University, Haikou 571199, China
| | - Rui Wang
- Key Laboratory of Emergency and Trauma, Ministry of Education, Key Laboratory of Hainan Trauma and Disaster Rescue, Key Laboratory of Haikou Trauma, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou 571199, China.
- Engineering Research Center for Hainan Bio-Smart Materials and Bio-Medical Devices, Key Laboratory of Hainan Functional Materials and Molecular Imaging, College of Pharmacy, College of Emergency and Trauma, Hainan Medical University, Haikou 571199, China
| | - Shiwei Li
- Key Laboratory of Emergency and Trauma, Ministry of Education, Key Laboratory of Hainan Trauma and Disaster Rescue, Key Laboratory of Haikou Trauma, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou 571199, China.
- Engineering Research Center for Hainan Bio-Smart Materials and Bio-Medical Devices, Key Laboratory of Hainan Functional Materials and Molecular Imaging, College of Pharmacy, College of Emergency and Trauma, Hainan Medical University, Haikou 571199, China
| | - Yanlong Xing
- Key Laboratory of Emergency and Trauma, Ministry of Education, Key Laboratory of Hainan Trauma and Disaster Rescue, Key Laboratory of Haikou Trauma, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou 571199, China.
- Engineering Research Center for Hainan Bio-Smart Materials and Bio-Medical Devices, Key Laboratory of Hainan Functional Materials and Molecular Imaging, College of Pharmacy, College of Emergency and Trauma, Hainan Medical University, Haikou 571199, China
| | - Fabiao Yu
- Key Laboratory of Emergency and Trauma, Ministry of Education, Key Laboratory of Hainan Trauma and Disaster Rescue, Key Laboratory of Haikou Trauma, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou 571199, China.
- Engineering Research Center for Hainan Bio-Smart Materials and Bio-Medical Devices, Key Laboratory of Hainan Functional Materials and Molecular Imaging, College of Pharmacy, College of Emergency and Trauma, Hainan Medical University, Haikou 571199, China
| |
Collapse
|
16
|
Li B, Tai Q, Yu H, Wang J, Gao M, Zhang X. Nanoliter Plasma Sample for Disease Diagnosis by Using a Visualization Platform of Counting Extracellular Vesicle Particles. Anal Chem 2025; 97:2786-2794. [PMID: 39895388 DOI: 10.1021/acs.analchem.4c04977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
Extracellular vesicles (EVs) are an excellent candidate for noninvasive diagnostic and prognostic assessments for liquid biopsy applications owing to reflecting the dynamic changes of pathological and physiological states in the body and exceptional stability in vivo. Herein, we developed a single-extracellular vesicle fluorescence visualization and counting platform containing three parts of the nanoliter-liquid operating platform for aspirating the sample, antibody-functionalized coverslip for capturing targeted EVs, and the single-molecule fluorescence labeling and imaging system for the visualization and counting of EVs. Compared to previously reported studies, our developed platform not only minimized operational complexity but also successfully detected single EVs using a minimal volume of 25 nL of plasma. Moreover, the results of fluorescence spot counts demonstrated that the developed platform exhibits a wide dynamic range and an excellent linear correlation between fluorescence intensity and particle numbers for the detection of CD9+ EVs from standard samples. Remarkably, our developed single EVs capture and analysis platform was successfully applied not only in the quantitative analysis of EpCAM+ EVs to CD9+ EVs for the diagnosis of breast cancer (BC) but also in the quantitative analysis of CD14+ EVs to CD9+ EVs for the diagnosis of type 2 diabetes (T2DM). Collectively, our work provides new ideas for the diagnosis of breast cancer and type 2 diabetes while developing a versatile method for distinguishing other diseases by simultaneous detection of multibiomarkers of Exos subtypes in trace samples due to the substitutability of antibodies.
Collapse
Affiliation(s)
- Bing Li
- Department of Chemistry and Institutes of Biomedical Sciences, Fudan University, Shanghai 200438, China
| | - Qunfei Tai
- Department of Chemistry and Institutes of Biomedical Sciences, Fudan University, Shanghai 200438, China
| | - Hailong Yu
- Department of Chemistry and Institutes of Biomedical Sciences, Fudan University, Shanghai 200438, China
| | - Jiaxi Wang
- The Human Phenome Institute, Zhangjiang-Fudan International Innovation Center, Fudan University, Shanghai 200438, China
| | - Mingxia Gao
- Department of Chemistry and Institutes of Biomedical Sciences, Fudan University, Shanghai 200438, China
| | - Xiangmin Zhang
- Department of Chemistry and Institutes of Biomedical Sciences, Fudan University, Shanghai 200438, China
| |
Collapse
|
17
|
Yang L, Zhang J, Zhang J, Hou T, Gao Q, Liu X, Li F. CRISPR-Based Homogeneous Electrochemical Strategy for Near-Zero Background Detection of Breast Cancer Extracellular Vesicles via Fluidity-Enhanced Magnetic Capture Nanoprobe. Anal Chem 2025; 97:2176-2185. [PMID: 39829104 DOI: 10.1021/acs.analchem.4c05181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
Precise identification and analysis of multiple protein biomarkers on the surface of breast cancer cell-derived extracellular vesicles (BC-EVs) are of great significance for noninvasive diagnosis of the breast cancer subtypes, but it remains a major challenge owing to their high heterogeneity and low abundance. Herein, we established a CRISPR-based homogeneous electrochemical strategy for near-zero background and ultrasensitive detection of BC-EVs. To realize the high-performance capture and isolation of BC-EVs, fluidity-enhanced magnetic nanoprobes were facilely prepared. After capturing BC-EVs, the AND logic gate-based catalytic hairpin assembly (CHA) and the trans-cleavage activity of CRISPR-Cas12a against the magnetic signal nanoprobes were triggered successively, generating a significant electrochemical signal. Notably, the as-developed metal-mediated magnetic signal nanoprobes could efficiently decrease the background signal by magnetic separation, endowing the method with a high signal-to-noise ratio. Consequently, by ingeniously integrating DNA logic gate-based CRISPR-CHA signal amplification with dual magnetic nanoprobes in a homogeneous electrochemical strategy, precise identification and ultrasensitive detection of BC-EVs was successfully achieved through simultaneous and specific recognition of dual protein markers on the BC-EVs surface. More importantly, this approach could effectively discriminate specific subgroups of BC-EVs in clinical serum samples, which may provide great opportunities for the accurate diagnosis and prognosis evaluation of breast cancer in a noninvasive manner.
Collapse
Affiliation(s)
- Limin Yang
- College of Chemistry and Pharmaceutical Sciences, Qingdao Agricultural University, Qingdao 266109, People's Republic of China
| | - Jingang Zhang
- College of Chemistry and Pharmaceutical Sciences, Qingdao Agricultural University, Qingdao 266109, People's Republic of China
| | - Jing Zhang
- Qingdao Hiser Hospital Affiliated of Qingdao University (Qingdao Traditional Chinese Medicine Hospital), Qingdao 266011, People's Republic of China
| | - Ting Hou
- College of Chemistry and Pharmaceutical Sciences, Qingdao Agricultural University, Qingdao 266109, People's Republic of China
| | - Qian Gao
- College of Chemistry and Pharmaceutical Sciences, Qingdao Agricultural University, Qingdao 266109, People's Republic of China
| | - Xiaojuan Liu
- College of Chemistry and Pharmaceutical Sciences, Qingdao Agricultural University, Qingdao 266109, People's Republic of China
| | - Feng Li
- College of Chemistry and Pharmaceutical Sciences, Qingdao Agricultural University, Qingdao 266109, People's Republic of China
| |
Collapse
|
18
|
Vancolen S, Chevin M, Robaire B, Sébire G. Exposure to Group B Streptococcus-induced chorioamnionitis alters the proteome of placental extracellular vesicles. Placenta 2025:S0143-4004(25)00018-9. [PMID: 39864996 DOI: 10.1016/j.placenta.2025.01.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 01/02/2025] [Accepted: 01/16/2025] [Indexed: 01/28/2025]
Abstract
INTRODUCTION Group B Streptococcus (GBS) is an opportunistic pathogen that can induce chorioamnionitis (CA), increasing the risk of neurodevelopmental disorders (NDDs) in the offspring. The placenta facilitates maternal-fetal communication through the release of extracellular vesicles (EVs), which may carry inflammatory molecules such as interleukin (IL)-1. Although the role of EVs in immune modulation is well established, their specific characterization in the context of GBS-induced CA has not yet been investigated. Understanding placental-derived EVs could further define how IL-1 and other inflammatory factors contribute to NDDs. METHODS We used an established rat model of GBS-induced CA. EVs from control and GBS infected dams were isolated from placentas and characterized using nanoparticle tracking analysis and transmission electron microscopy. The protein content was assessed via mass spectrometry, followed by subsequent pathway analysis. ELISA was used to quantify cytokine levels. RESULTS GBS-infected placentas exhibited calcification and increased weight, while fetal weight decreased. Analysis of the proteome from control versus GBS placental EVs revealed distinct profiles, with many proteins involved in the innate immune response, including alarmins (S100A8/9), complement pathways, and cytokine signaling pathways. Pathway analysis highlighted IL-1α and IL-1β identified as key upstream regulators. Notably, EVs from GBS-infected males showed a 44-fold increase in intracellular IL-1β compared to controls. DISCUSSION These findings indicate that GBS-induced CA alters the protein content of EVs from placental cells. Our findings of increased IL-1β-associated EVs highlight the need for further investigation into the role of these cytokines from GBS-exposed placentas and their role in brain injuries leading to NDDs.
Collapse
Affiliation(s)
- Seline Vancolen
- Department of Pharmacology & Therapeutics, McGill University, Montreal, Quebec, Canada; Department of Pediatrics, Research Institute of the McGill University Health Center, Montreal, Quebec, Canada
| | - Mathilde Chevin
- Department of Pediatrics, Research Institute of the McGill University Health Center, Montreal, Quebec, Canada
| | - Bernard Robaire
- Department of Pharmacology & Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Guillaume Sébire
- Department of Pediatrics, Research Institute of the McGill University Health Center, Montreal, Quebec, Canada.
| |
Collapse
|
19
|
Fan R, Chen S, Lan F, Li W, Zhu Y, Zhang L, Zhang Y, Li L. Surface-Enhanced Raman Scattering (SERS)-based biosensors for advanced extracellular vesicle detection: A review. Anal Chim Acta 2025; 1336:343264. [PMID: 39788643 DOI: 10.1016/j.aca.2024.343264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 09/18/2024] [Accepted: 09/19/2024] [Indexed: 01/12/2025]
Abstract
BACKGROUND Extracellular Vesicles (EVs), as nano-scale vesicles rich in biological information, hold an indispensable status in the biomedical field. However, due to the intrinsic small size and low abundance of EVs, their effective detection presents significant challenges. Although various EV detection techniques exist, their sensitivity and ease of operation still need enhancement. RESULTS Surface-Enhanced Raman Scattering (SERS) is known for its high sensitivity and specificity. It stands out in tackling the challenges that traditional EV detection methods face. In this review, we focus on the application of SERS-based biosensors in EV detection. It provides a detailed introduction to the recognition and capture of EVs, strategies for mediating signal amplification, and detection of EV biomarkers. Finally, the challenges and prospects of SERS-based biosensors are discussed. SIGNIFICANCE SERS-based biosensor enhances the Raman signal, allowing for the detection of biomarkers at low concentrations. This capability reveals its substantial potential in identifying EVs and analyzing molecular data. It paves the path for advanced EV detection.
Collapse
Affiliation(s)
- Rui Fan
- School of Medical Technology, Guangdong Medical University, Dongguan, 523808, China; 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, PR China
| | - Siting Chen
- 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, PR China; School of Basic Medical Science, Southern Medical University, Guangzhou, 510515, China
| | - Fei Lan
- 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, PR China
| | - Wenbin Li
- 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, PR China
| | - Yitong Zhu
- 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, PR China; School of Basic Medical Science, Southern Medical University, Guangzhou, 510515, China
| | - Lifeng Zhang
- School of Medical Technology, Guangdong Medical University, Dongguan, 523808, China; 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, PR China
| | - Ye Zhang
- 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, PR China.
| | - Ling Li
- School of Medical Technology, Guangdong Medical University, Dongguan, 523808, China; School of Basic Medical Science, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
20
|
Lica JJ, Jakóbkiewicz-Banecka J, Hellmann A. In Vitro models of leukemia development: the role of very small leukemic stem-like cells in the cellular transformation cascade. Front Cell Dev Biol 2025; 12:1463807. [PMID: 39830209 PMCID: PMC11740207 DOI: 10.3389/fcell.2024.1463807] [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: 07/15/2024] [Accepted: 11/28/2024] [Indexed: 01/22/2025] Open
Abstract
Recent experimental findings indicate that cancer stem cells originate from transformed very small embryonic-like stem cells. This finding represents an essential advancement in uncovering the processes that drive the onset and progression of cancer. In continuously growing cell lines, for the first time, our team's follow-up research on leukemia, lung cancer, and healthy embryonic kidney cells revealed stages that resembles very small precursor stem cells. This review explores the origin of leukemic stem-like cells from very small leukemic stem-like cells establish from transformed very small embryonic-like stem cells. We explore theoretical model of acute myeloid leukemia initiation and progresses through various stages, as well basing the HL60 cell line, present its hierarchical stage development in vitro, highlighting the role of these very small precursor primitive stages. We also discuss the potential implications of further research into these unique cellular stages for advancing leukemia and cancer treatment and prevention.
Collapse
Affiliation(s)
- Jan Jakub Lica
- Department Medical Biology and Genetics, Faculty of Biology, University of Gdansk, Gdansk, Poland
- Department Health Science; Powiśle University, Gdańsk, Poland
| | | | - Andrzej Hellmann
- Department of Hematology and Transplantology, Faculty of Medicine, Medical University of Gdansk, Gdańsk, Poland
| |
Collapse
|
21
|
Kwaku GN, Jensen KN, Simaku P, Floyd DJ, Saelens JW, Reardon CM, Ward RA, Basham KJ, Hepworth OW, Vyas TD, Zamith-Miranda D, Nosanchuk JD, Vyas JM, Harding HB. Extracellular vesicles from diverse fungal pathogens induce species-specific and endocytosis-dependent immunomodulation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.03.631181. [PMID: 39803513 PMCID: PMC11722428 DOI: 10.1101/2025.01.03.631181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/21/2025]
Abstract
Microbial pathogens generate extracellular vesicles (EVs) for intercellular communication and quorum sensing. Microbial EVs also induce inflammatory pathways within host innate immune cells. We previously demonstrated that EVs secreted by Candida albicans trigger type I interferon signaling in host cells specifically via the cGAS-STING innate immune signaling pathway. Here, we show that despite sharing similar properties of morphology and internal DNA content, the interactions between EVs and the innate immune system differ according to the parental fungal species. EVs secreted by C. albicans, Saccharomyces cerevisiae, Cryptococcus neoformans, and Aspergillus fumigatus are endocytosed at different rates by murine macrophages triggering varied cytokine responses, innate immune signaling, and subsequent immune cell recruitment. Notably, cell wall constituents that decorate C. neoformans and A. fumigatus EVs inhibit efficient internalization by macrophages and dampen innate immune activation. Our data uncover the transcriptional and functional consequences of the internalization of diverse fungal EVs by immune cells and reveal novel insights into the early innate immune response to distinct clinically significant fungal pathogens.
Collapse
Affiliation(s)
- Geneva N Kwaku
- Division of Infectious Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Kirstine Nolling Jensen
- Division of Infectious Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Patricia Simaku
- Division of Infectious Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Daniel J Floyd
- Division of Infectious Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Joseph W Saelens
- Pfizer Worldwide Research Development and Medical, Machine Learning and Computational Sciences, Cambridge, MA, USA
| | - Christopher M Reardon
- Division of Infectious Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Rebecca A Ward
- Division of Infectious Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Kyle J Basham
- Division of Infectious Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Olivia W Hepworth
- Division of Infectious Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Tammy D Vyas
- Division of Infectious Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Daniel Zamith-Miranda
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, USA
- Division of Infectious Diseases, Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Joshua D Nosanchuk
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, USA
- Division of Infectious Diseases, Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Jatin M Vyas
- Division of Infectious Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Division of Infectious Disease, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
| | - Hannah Brown Harding
- Division of Infectious Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| |
Collapse
|
22
|
Fowler SL, Behr TS, Turkes E, O'Brien DP, Cauhy PM, Rawlinson I, Edmonds M, Foiani MS, Schaler A, Crowley G, Bez S, Ficulle E, Tsefou E, Fischer R, Geary B, Gaur P, Miller C, D'Acunzo P, Levy E, Duff KE, Ryskeldi-Falcon B. Tau filaments are tethered within brain extracellular vesicles in Alzheimer's disease. Nat Neurosci 2025; 28:40-48. [PMID: 39572740 PMCID: PMC11706778 DOI: 10.1038/s41593-024-01801-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 09/25/2024] [Indexed: 11/27/2024]
Abstract
The abnormal assembly of tau protein in neurons is a pathological hallmark of multiple neurodegenerative diseases, including Alzheimer's disease (AD). Assembled tau associates with extracellular vesicles (EVs) in the central nervous system of individuals with AD, which is linked to its clearance and prion-like propagation. However, the identities of the assembled tau species and EVs, as well as how they associate, are not known. Here, we combined quantitative mass spectrometry, cryo-electron tomography and single-particle cryo-electron microscopy to study brain EVs from individuals with AD. We found tau filaments composed mainly of truncated tau that were enclosed within EVs enriched in endo-lysosomal proteins. We observed multiple filament interactions, including with molecules that tethered filaments to the EV limiting membrane, suggesting selective packaging. Our findings will guide studies into the molecular mechanisms of EV-mediated secretion of assembled tau and inform the targeting of EV-associated tau as potential therapeutic and biomarker strategies for AD.
Collapse
Affiliation(s)
- Stephanie L Fowler
- UK Dementia Research Institute at University College London, London, UK
- Oxford-GSK Institute of Molecular and Computational Medicine, University of Oxford, Oxford, UK
| | - Tiana S Behr
- Medical Research Council Laboratory of Molecular Biology, Cambridge, UK
| | - Emir Turkes
- UK Dementia Research Institute at University College London, London, UK
| | - Darragh P O'Brien
- Target Discovery Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | | | - Isadora Rawlinson
- Medical Research Council Laboratory of Molecular Biology, Cambridge, UK
| | - Marisa Edmonds
- UK Dementia Research Institute at University College London, London, UK
| | - Martha S Foiani
- UK Dementia Research Institute at University College London, London, UK
| | - Ari Schaler
- Taub Institute, Irving Medical Center, Columbia University, New York, NY, USA
| | - Gerard Crowley
- UK Dementia Research Institute at University College London, London, UK
| | - Sumi Bez
- UK Dementia Research Institute at University College London, London, UK
| | - Elena Ficulle
- UK Dementia Research Institute at University College London, London, UK
| | - Eliona Tsefou
- UK Dementia Research Institute at University College London, London, UK
| | - Roman Fischer
- Target Discovery Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Beth Geary
- Medical Research Council Protein Phosphorylation and Ubiquitylation Unit, University of Dundee, Dundee, UK
| | - Pallavi Gaur
- Department of Neurology, Center for Translational and Computational Neuroimmunology, Columbia University Medical Center, New York, NY, USA
| | - Chelsea Miller
- The Center for Genetic and Genomic Medicine, Hackensack University Medical Center, Hackensack, NJ, USA
- Center for Dementia Research, Nathan S. Kline Institute for Psychiatric Research, Orangeburg, NY, USA
| | - Pasquale D'Acunzo
- Center for Dementia Research, Nathan S. Kline Institute for Psychiatric Research, Orangeburg, NY, USA
- Department of Psychiatry, New York University Grossman School of Medicine, New York, NY, USA
| | - Efrat Levy
- Center for Dementia Research, Nathan S. Kline Institute for Psychiatric Research, Orangeburg, NY, USA
- Department of Psychiatry, New York University Grossman School of Medicine, New York, NY, USA
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, New York, NY, USA
- Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, USA
| | - Karen E Duff
- UK Dementia Research Institute at University College London, London, UK.
- Taub Institute, Irving Medical Center, Columbia University, New York, NY, USA.
| | | |
Collapse
|
23
|
Qin X, Hu KL, Li Q, Sun Y, Peng T, Liu X, Li J, Nan W, Yu Y, Qi X, Li R. In Situ Sprayed Hydrogel Delivers Extracellular Vesicles Derived from Human Endometrial Organoids for Uterine Function Preservation and Fertility Restoration. Adv Healthc Mater 2025; 14:e2403604. [PMID: 39558805 DOI: 10.1002/adhm.202403604] [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: 09/20/2024] [Indexed: 11/20/2024]
Abstract
Impaired endometrial function and reduced receptivity remain significant causes of female infertility. Here, a sprayable hydrogel combined with human endometrial organoid extracellular vesicles (HEO-EVs) is developed to enhance uterine function preservation and fertility restoration. The peptide amphiphile hydrogel (labeled CPA) is engineered by conjugating a collagen-binding peptide with glutathione to impart its biocompatible adhesive and antioxidant properties. The therapeutic EVs are isolated and purified from human endometrial organoids that have been stably passaged long-term using a bioreactor-culture system. The resulting HEO-EVs-loaded CPA (CPA@HEO-EVs) rapid gelation, triggered by salt-ion interactions, occurs when the fluid is sprayed onto the uterine lining. The ex vivo studies demonstrate that CPA@HEO-EVs promote cell proliferation, scavenges free radicals, and increases tube formation in human umbilical vein endothelial cells. In vivo experiments further validate that in situ spraying with the CPA@HEO-EVs can promote neovascularization, prevent localized endometrial fibrosis, and effectively enhance fertility in a mouse model of endometrial injury. These findings highlight the promising clinical application of in situ sprayed CPA@HEO-EVs hydrogel for targeted endometrial therapy.
Collapse
Affiliation(s)
- Xunsi Qin
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, No. 49 North Huayuan Road, Haidian District, Beijing, 100191, China
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, 100191, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive, Technology, Beijing, 100191, China
| | - Kai-Lun Hu
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, No. 49 North Huayuan Road, Haidian District, Beijing, 100191, China
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, 100191, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive, Technology, Beijing, 100191, China
| | - Qi Li
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, No. 49 North Huayuan Road, Haidian District, Beijing, 100191, China
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, 100191, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive, Technology, Beijing, 100191, China
| | - Yuze Sun
- National Institute of Biological Sciences, Zhongguancun Life Science Park, 7 Science Park Road, Beijing, 102206, China
- Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing, 100084, China
| | - Tianliu Peng
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, No. 49 North Huayuan Road, Haidian District, Beijing, 100191, China
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, 100191, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive, Technology, Beijing, 100191, China
| | - Xiyao Liu
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, No. 49 North Huayuan Road, Haidian District, Beijing, 100191, China
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, 100191, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive, Technology, Beijing, 100191, China
| | - Jizhou Li
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, No. 49 North Huayuan Road, Haidian District, Beijing, 100191, China
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, 100191, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive, Technology, Beijing, 100191, China
| | - Wenhui Nan
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, No. 49 North Huayuan Road, Haidian District, Beijing, 100191, China
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, 100191, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive, Technology, Beijing, 100191, China
| | - Yang Yu
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, No. 49 North Huayuan Road, Haidian District, Beijing, 100191, China
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, 100191, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive, Technology, Beijing, 100191, China
| | - Xiangbing Qi
- National Institute of Biological Sciences, Zhongguancun Life Science Park, 7 Science Park Road, Beijing, 102206, China
- Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing, 100084, China
| | - Rong Li
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, No. 49 North Huayuan Road, Haidian District, Beijing, 100191, China
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, 100191, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive, Technology, Beijing, 100191, China
| |
Collapse
|
24
|
Chen Q, Li Y, Hu J, Xu Z, Wang S, Cai N, He M, Xiao Y, Ding Y, Sun M, Li C, Cao Y, Wang Z, Zhou F, Wang G, Wang C, Tu J, Hu H, Sun C. Local Exosome Inhibition Potentiates Mild Photothermal Immunotherapy Against Breast Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2406328. [PMID: 39574346 PMCID: PMC11727390 DOI: 10.1002/advs.202406328] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 11/12/2024] [Indexed: 01/14/2025]
Abstract
Limited immune infiltration within the tumor microenvironment (TME) hampers the efficacy of immune checkpoint blockade (ICB) therapy. To enhance immune infiltration, mild photothermal therapy (PTT) is often combined with immunotherapy. However, the impact of mild PTT on the TME remains unclear. The bioinformatics analyses reveal that mild PTT amplifies immune cell infiltration and stimulates T-cell activity. Notably, it accelerates the release of tumor cell-derived exosomes (TEX) and upregulates PD-L1 expression on both tumor cells and TEX. Consequently, it is proposed that locally inhibiting TEX release is crucial for overcoming the adverse effects of mild PTT, thereby enhancing ICB therapy. Thus, a multi-stage drug delivery system is designed that concurrently delivers photosensitizers (reduced graphene oxide nanosheets, NRGO), anti-PD-L1 antibodies, and exosome inhibitors (sulfisoxazole). The system employs a temperature-sensitive lipid gel as the primary carrier, with NRGO serving as a secondary carrier that supports photothermal conversion and incorporation of sulfisoxazole. Importantly, controlled drug release is achieved using near-infrared radiation. The findings indicate that this local combination therapy remodels the immunosuppressive TME through exosome inhibition and enhanced immune cell infiltration, while also boosting T-cell activity to trigger systemic antitumor immunity, showcasing the remarkable efficacy of this combination strategy in eradicating cold tumors.
Collapse
|
25
|
Perera N, De Blasio MJ, Febbraio MA. Harnessing the therapeutic potential of exercise in extracellular vesicle-based therapy in metabolic disease associated cardiovascular complications. Free Radic Biol Med 2025; 226:230-236. [PMID: 39549882 DOI: 10.1016/j.freeradbiomed.2024.11.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Revised: 10/10/2024] [Accepted: 11/13/2024] [Indexed: 11/18/2024]
Abstract
Cardiovascular disease (CVD) is a leading cause of mortality, affecting ∼18 million individuals each year. Obesity and type 2 diabetes mellitus in particular, both chronic metabolic disorders, are risk factors for CVD. The salutary effects of physical activity in preventing and ameliorating CVD have long been acknowledged, as it improves glucose and lipid homeostasis, alongside attenuating oxidative damage, increasing mitochondrial function, and ultimately improving cardiac function. Exercise serves as a catalyst for the secretion of extracellular vesicles (EVs), facilitating inter-tissue communication, by which tissues can deliver important signals from one tissue to another. In recent years, an increasing number of studies have focused on the cargo encapsulated within exercise-derived EVs, as well as the orchestration of inter-tissue crosstalk aimed at modulating metabolism and tissue function in CVDs. The precise mechanisms underpinning the cardioprotective properties of exercise-derived EVs, however, remains only partially elucidated. This review explores novel EV based therapeutic options in CVD and, in particular, EVs derived from models of exercise to alter metabolism and enhance cardiovascular outcomes.
Collapse
Affiliation(s)
- Nimna Perera
- Monash Institute of Pharmaceutical Sciences, Parkville, Melbourne, Australia
| | - Miles J De Blasio
- Monash Institute of Pharmaceutical Sciences, Parkville, Melbourne, Australia
| | - Mark A Febbraio
- Monash Institute of Pharmaceutical Sciences, Parkville, Melbourne, Australia.
| |
Collapse
|
26
|
Bergqvist M, Lässer C, Crescitelli R, Park K, Lötvall J. A Non-Centrifugation Method to Concentrate and Purify Extracellular Vesicles Using Superabsorbent Polymer Followed by Size Exclusion Chromatography. J Extracell Vesicles 2025; 14:e70037. [PMID: 39840900 PMCID: PMC11752139 DOI: 10.1002/jev2.70037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Accepted: 12/27/2024] [Indexed: 01/23/2025] Open
Abstract
Extracellular vesicles (EVs) can be isolated and purified from cell cultures and biofluids using different methodologies. Here, we explored a novel EV isolation approach by combining superabsorbent polymers (SAP) in a dialysis membrane with size exclusion chromatography (SEC) to achieve high concentration and purity of EVs without the use of ultracentrifugation (UC). Suspension HEK293 cells transfected with CD63 coupled with Thermo Luciferase were used to quantify the EV yield and purity. The 500 mL conditioned medium volume was initially reduced by pressure ultrafiltration, followed by UC, SAP or a centrifugal filter unit (CFU). Using either of these methods, the EVs were concentrated to a final volume of approximately 1 mL, with retained functionality. The yield, quantified by luciferase activity, was highest with UC (70%-80%), followed by SAP (60%-70%) and CFU (50%-60%). Further purification of the EVs was performed by iodixanol density cushion (IDC) or SEC (Sepharose CL-2B or 6B, in either 10 or 20 mL columns). Although the IDC and Sepharose CL-2B (10 mL) achieved the highest yields, the purity was slightly higher (30%) with IDC. In conclusion, combining SAP concentration with CL-2B SEC is an alternative and efficient way to isolate EVs without using UC.
Collapse
Affiliation(s)
- Markus Bergqvist
- Department of Internal Medicine and Clinical Nutrition, Krefting Research Centre, Institute of Medicine, Sahlgrenska Academy, University of GothenburgGothenburgSweden
| | - Cecilia Lässer
- Department of Internal Medicine and Clinical Nutrition, Krefting Research Centre, Institute of Medicine, Sahlgrenska Academy, University of GothenburgGothenburgSweden
| | - Rossella Crescitelli
- Department of Surgery, Sahlgrenska Center for Cancer Research and Wallenberg Centre for Molecular and Translational Medicine, Institute of Clinical Sciences, Sahlgrenska Academy, University of GothenburgGothenburgSweden
| | - Kyong‐Su Park
- Department of Internal Medicine and Clinical Nutrition, Krefting Research Centre, Institute of Medicine, Sahlgrenska Academy, University of GothenburgGothenburgSweden
| | - Jan Lötvall
- Department of Internal Medicine and Clinical Nutrition, Krefting Research Centre, Institute of Medicine, Sahlgrenska Academy, University of GothenburgGothenburgSweden
| |
Collapse
|
27
|
Lorite P, Domínguez JN, Palomeque T, Torres MI. Extracellular Vesicles: Advanced Tools for Disease Diagnosis, Monitoring, and Therapies. Int J Mol Sci 2024; 26:189. [PMID: 39796048 PMCID: PMC11720073 DOI: 10.3390/ijms26010189] [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: 11/29/2024] [Revised: 12/23/2024] [Accepted: 12/26/2024] [Indexed: 01/13/2025] Open
Abstract
Extracellular vesicles (EVs) are a heterogeneous group of membrane-encapsulated vesicles released by cells into the extracellular space. They play a crucial role in intercellular communication by transporting bioactive molecules such as proteins, lipids, and nucleic acids. EVs can be detected in body fluids, including blood plasma, urine, saliva, amniotic fluid, breast milk, and pleural ascites. The complexity and diversity of EVs require a robust and standardized approach. By adhering to standardized protocols and guidelines, researchers can ensure the consistency, purity, and reproducibility of isolated EVs, facilitating their use in diagnostics, therapies, and research. Exosomes and microvesicles represent an exciting frontier in modern medicine, with significant potential to transform the diagnosis and treatment of various diseases with an important role in personalized medicine and precision therapy. The primary objective of this review is to provide an updated analysis of the significance of EVs by highlighting their mechanisms of action and exploring their applications in the diagnosis and treatment of various diseases. Additionally, the review addresses the existing limitations and future potential of EVs, offering practical recommendations to resolve current challenges and enhance their viability for clinical use. This comprehensive approach aims to bridge the gap between EV research and its practical application in healthcare.
Collapse
Affiliation(s)
| | | | | | - María Isabel Torres
- Department of Experimental Biology, Faculty of Health Sciences, University of Jaén, 23071 Jaén, Spain; (P.L.); (J.N.D.); (T.P.)
| |
Collapse
|
28
|
Shi S, Liu X, Geng X, Meng Q, Gao M, Wang E, Ma X, Hu H, Liu J, Han W, Yin H, Zhou X. Neonatal heart tissue-derived EVs alleviate adult ischemic cardiac injury via regulating the function of macrophages and cardiac regeneration in murine models. Int Immunopharmacol 2024; 143:113251. [PMID: 39353386 DOI: 10.1016/j.intimp.2024.113251] [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: 05/22/2024] [Revised: 09/04/2024] [Accepted: 09/22/2024] [Indexed: 10/04/2024]
Abstract
Previous studies confirmed the regenerative capacity of the mammalian neonatal heart. We recently found that adult heart tissue-derived EVs can protect the heart from myocardial ischemia-reperfusion (I/R). However, the role of EVs from neonatal heart tissue in cardiac healing post-ischemia remains unclear. In the present study, we revealed that intramyocardial administration of neonatal cardiac tissue-derived EVs (ncEVs) alleviated cardiac inflammation, mitigated reperfusion injury, and improved cardiac function in murine I/R models. In vitro, ncEVs inhibited M1 polarization of macrophages induced by LPS while up-regulated their phagocytic function via the miR-133a-3p-Ash1l signaling pathway. Moreover, the administration of ncEVs contributed to cardiac angiogenesis and improved cardiac function in murine myocardial infarction models. Collectively, these results suggested that neonatal heart-derived EVs can regulate the function of macrophages and contribute to cardiac regeneration and function recovery in murine cardiac ischemic models. Therefore, the derivatives in neonatal heart tissue-derived EVs might serve as a potential therapeutic strategy in ischemic diseases.
Collapse
Affiliation(s)
- Shanshan Shi
- Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China; Shanghai Heart Failure Research Center, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China; Department of Pathology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Xuan Liu
- Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China; Shanghai Heart Failure Research Center, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China; Department of Cardiothoracic Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Xuedi Geng
- Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China; Shanghai Heart Failure Research Center, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Qingshu Meng
- Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China; Shanghai Heart Failure Research Center, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Mingkui Gao
- Department of Cardiothoracic Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Enhao Wang
- Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China; Shanghai Heart Failure Research Center, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Xiaoxue Ma
- Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China; Shanghai Heart Failure Research Center, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Hao Hu
- Department of Heart Failure, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China; Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai 200120, China
| | - Jie Liu
- Department of Thoracic Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Wei Han
- Department of Heart Failure, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China; Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai 200120, China
| | - Hui Yin
- Department of Thoracic Surgery, The First Affiliated Hospital of Shaoyang University, Shaoyang 422000 China.
| | - Xiaohui Zhou
- Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China; Shanghai Heart Failure Research Center, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China.
| |
Collapse
|
29
|
Zhu Q, Mao X, Zhu X, Xiao Y, Xu H, Su L, Liu X, Huang X, Wang L. Hypoxia-Induced and Glucuronic Acid-Modified Extracellular Vesicles from Mesenchymal Stromal Cells Treat Pulmonary Arterial Hypertension by Improving Vascular Remodeling. NANO LETTERS 2024; 24:16342-16350. [PMID: 39660764 DOI: 10.1021/acs.nanolett.4c04638] [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: 12/12/2024]
Abstract
Achieving precise delivery of extracellular vesicles (EVs) to treat pulmonary arterial hypertension (PAH) remains challenging. Here, we propose a strategy using hypoxia-induced and glucuronic acid (GA)-modified mesenchymal stromal-cell-derived EVs (MSC-EVs) to enhance their functionalities and therapeutic targeting. The hypoxia-induced EVs (Hypo-EVs) exhibit enriched exosomal signatures and display heightened inhibition of the proliferation of pulmonary arterial smooth muscle cells (PASMCs) compared to normoxic EVs (Norm-EV). We then modify Hypo-EVs by incorporating GA into their outer membrane, targeting glucose transporter-1 overexpressed on PASMCs. Our studies show that GA-EVs significantly enhance the therapeutic efficacy, both in vitro and in vivo, through improved targeted delivery to diseased PASMCs for improving vascular remodeling. Additionally, we identify miR-5119 involved in the PAH-associated calcium signaling pathway as a key contributor to GA-EVs' superior effects. This work provides a promising strategy for PAH treatment and advances the clinical potential of MSC-EV-based therapies.
Collapse
Affiliation(s)
- Qingfu Zhu
- National Engineering Research Center of Ophthalmology and Optometry, School of Biomedical Engineering, Eye Hospital, Wenzhou Medical University,Xueyuan Road 270, Wenzhou 325027, China
| | - Xulong Mao
- Division of Pulmonary Medicine, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
- Wenzhou Key Laboratory of Interdiscipline and Translational Medicine, Wenzhou Key Laboratory of Heart and Lung, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Xinxi Zhu
- Division of Pulmonary Medicine, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
- Wenzhou Key Laboratory of Interdiscipline and Translational Medicine, Wenzhou Key Laboratory of Heart and Lung, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Yijia Xiao
- Division of Pulmonary Medicine, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
- Wenzhou Key Laboratory of Interdiscipline and Translational Medicine, Wenzhou Key Laboratory of Heart and Lung, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Hao Xu
- National Engineering Research Center of Ophthalmology and Optometry, School of Biomedical Engineering, Eye Hospital, Wenzhou Medical University,Xueyuan Road 270, Wenzhou 325027, China
| | - Lihuang Su
- Division of Pulmonary Medicine, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
- Wenzhou Key Laboratory of Interdiscipline and Translational Medicine, Wenzhou Key Laboratory of Heart and Lung, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Xiaohu Liu
- National Engineering Research Center of Ophthalmology and Optometry, School of Biomedical Engineering, Eye Hospital, Wenzhou Medical University,Xueyuan Road 270, Wenzhou 325027, China
| | - Xiaoying Huang
- Division of Pulmonary Medicine, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
- Wenzhou Key Laboratory of Interdiscipline and Translational Medicine, Wenzhou Key Laboratory of Heart and Lung, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Liangxing Wang
- Division of Pulmonary Medicine, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
- Wenzhou Key Laboratory of Interdiscipline and Translational Medicine, Wenzhou Key Laboratory of Heart and Lung, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| |
Collapse
|
30
|
Li X, Qi X, Liu X, Zhu J, Hu L. Lipopolysaccharide Imprinted Polymers for Specific Recognition of Bacterial Outer Membrane Vesicles. Anal Chem 2024; 96:19803-19811. [PMID: 39572928 DOI: 10.1021/acs.analchem.4c05288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2024]
Abstract
Outer membrane vesicles (OMVs) secreted by bacteria are emerging diagnostic markers for bacterial infection or disease detection due to their carriage of various signaling molecules. However, actual biological samples of patients are extremely complex, and applying OMVs to clinical diagnosis remains a major challenge. One of the major challenges is that there are still great difficulties in the enrichment of OMVs including tedious steps and lower concentration. And some commonly used exosome enrichment methods, such as ultracentrifugation, still have some shortcomings. Herein, we introduce lipopolysaccharide (LPS) molecularly imprinted polymer (MIP) for efficient capturing and analyzing OMVs, enabling a novel approach to bacterial disease diagnosis based on biorecognition materials. LPS, as a unique structure of Gram-negative bacteria, also widely expressed on the surface of OMVs, which will form cyclic hydrogen bonds with functional monomers of MIP with affinity interactions. The prepared MIP efficiently can isolate OMVs from 100 μL of culture broth via specific affinity LPS in less than 40 min with a recovery rate of over 95%. Moreover, MIP exhibits good reusability, with almost identical enrichment performance after 5 repeated cycles, contributing to reducing experimental costs in both time and economy. The captured OMVs can be detected using Western blotting with target protein antibodies or in combination with proteomic analysis, providing a proteomic biomarker platform for early bacteria disease diagnosis.
Collapse
Affiliation(s)
- Xiaojun Li
- Center for Supramolecular Chemical Biology, State Key Laboratory of Supramolecular Structure and Materials, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Xiulei Qi
- Center for Supramolecular Chemical Biology, State Key Laboratory of Supramolecular Structure and Materials, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Xingguo Liu
- Center for Supramolecular Chemical Biology, State Key Laboratory of Supramolecular Structure and Materials, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Jun Zhu
- Jingjie PTM BioLab Co. Ltd., Hangzhou 310018, China
| | - Lianghai Hu
- Center for Supramolecular Chemical Biology, State Key Laboratory of Supramolecular Structure and Materials, School of Life Sciences, Jilin University, Changchun 130012, China
| |
Collapse
|
31
|
Cuellar-Gaviria TZ, Rincon-Benavides MA, Halipci Topsakal HN, Salazar-Puerta AI, Jaramillo-Garrido S, Kordowski M, Vasquez-Martinez CA, Nguyen KT, Rima XY, Rana PSJB, Combita-Heredia O, Deng B, Dathathreya K, McComb DW, Reategui E, Wozniak D, Higuita-Castro N, Gallego-Perez D. Tissue nano-transfection of antimicrobial genes drives bacterial biofilm killing in wounds and is potentially mediated by extracellular vesicles. J Control Release 2024; 376:1300-1315. [PMID: 39491627 PMCID: PMC11780627 DOI: 10.1016/j.jconrel.2024.10.071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Revised: 10/06/2024] [Accepted: 10/30/2024] [Indexed: 11/05/2024]
Abstract
The emergence of bacteria that are resistant to antibiotics is on track to become a major global health crisis. Therefore, there is an urgent need for new treatment options. Here, we studied the implementation of tissue-nanotransfection (TNT) to treat Staphylococcus aureus-infected wounds by delivering gene cargos that boost the levels of naturally produced antimicrobial peptides. The Cathelicidin Antimicrobial Peptide gene (CAMP), which produces the antimicrobial peptide LL-37, was used as model gene cargo. In vitro evaluation showed successful transfection and an increase in the transcription and translation of CAMP-coding plasmid in mouse primary epithelial cells. Moreover, we found that the extracellular vesicles (EVs) derived from the transfected cells (in vitro and in vivo) carried significantly higher concentrations of CAMP transcripts and LL-37 peptide compared to control EVs, possibly mediating the trafficking of the antimicrobial contents to other neighboring cells. The TNT platform was then used in vivo on an excisional wound model in mice to nanotransfect the CAMP-coding plasmid on the edge of infected wounds. After 4 days of daily treatment, we observed a significant decrease in the bacterial load in the CAMP-treated group compared to the sham group. Moreover, histological analysis and bacterial load quantification also revealed that TNT of CAMP on S. aureus-infected wounds was effective in treating biofilm progression by reducing the bacterial load. Lastly, we observed a significant increase in macrophage recruitment to the infected tissue, a robust increase in vascularization, as well as and an increased expression of IL10 and Fli1. Our results demonstrate that TNT-based delivery of gene cargos coding for antimicrobial compounds to the wound is a promising approach for combating biofilm infections in wounds.
Collapse
Affiliation(s)
- Tatiana Z Cuellar-Gaviria
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA; Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA; Gene Therapy Institute, The Ohio State University, Columbus, OH 43210, USA; Infectious Disease Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Maria Angelica Rincon-Benavides
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA; Biophysics Program, The Ohio State University, Columbus, OH 43210, USA
| | - Hatice Nur Halipci Topsakal
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA; Istanbul Atlas University, Istanbul 34408, Turkiye
| | | | | | - Mia Kordowski
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA; Biophysics Program, The Ohio State University, Columbus, OH 43210, USA
| | - Carlos A Vasquez-Martinez
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA; CONACYT - Faculty of Medicine, Benito Juárez Autonomous University of Oaxaca, Oaxaca 68020, Mexico
| | - Kim Truc Nguyen
- William G. Lowrie Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH 43210, USA
| | - Xilal Y Rima
- William G. Lowrie Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH 43210, USA
| | - Pranav S J B Rana
- Department of Microbiology, The Ohio State University, Columbus, OH 43210, USA; Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH 43210, USA
| | | | - Binbin Deng
- Center for Electron Microscopy and Analysis (CEMAS), The Ohio State University, Columbus, OH 43210, USA
| | - Kavya Dathathreya
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA
| | - David W McComb
- Center for Electron Microscopy and Analysis (CEMAS), The Ohio State University, Columbus, OH 43210, USA; Department of Materials Science and Engineering, The Ohio State University, Columbus, OH 43210, USA
| | - Eduardo Reategui
- William G. Lowrie Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH 43210, USA; Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| | - Daniel Wozniak
- Infectious Disease Institute, The Ohio State University, Columbus, OH 43210, USA; Department of Microbiology, The Ohio State University, Columbus, OH 43210, USA; Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH 43210, USA
| | - Natalia Higuita-Castro
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA; Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA; Gene Therapy Institute, The Ohio State University, Columbus, OH 43210, USA; Infectious Disease Institute, The Ohio State University, Columbus, OH 43210, USA; Biophysics Program, The Ohio State University, Columbus, OH 43210, USA; Department of Neurological Surgery, The Ohio State University, Columbus, OH 43210, USA
| | - Daniel Gallego-Perez
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA; Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA; Gene Therapy Institute, The Ohio State University, Columbus, OH 43210, USA; Infectious Disease Institute, The Ohio State University, Columbus, OH 43210, USA; Biophysics Program, The Ohio State University, Columbus, OH 43210, USA; Department of Surgery, The Ohio State University, Columbus, OH 43210, USA.
| |
Collapse
|
32
|
Jiang P, Ma X, Wang X, Huang J, Wang Y, Ai J, Xiao H, Dai M, Lin Y, Shao B, Tang X, Tong W, Ye Z, Chai R, Zhang S. Isolation and Comprehensive Analysis of Cochlear Tissue-Derived Small Extracellular Vesicles. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2408964. [PMID: 39497619 DOI: 10.1002/advs.202408964] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 10/19/2024] [Indexed: 12/28/2024]
Abstract
Small extracellular vesicles (sEVs) act as a critical mediator in intercellular communication. Compared to sEVs derived from in vitro sources, tissue-derived sEVs can reflect the in vivo signals released from specific tissues more accurately. Currently, studies on the role of sEVs in the cochlea have relied on studying sEVs from in vitro sources. This study evaluates three cochlear tissue digestion and cochlear tissue-derived sEV (CDsEV) isolation methods, and first proposes that the optimal approach for isolating CDsEVs using collagenase D and DNase І combined with sucrose density gradient centrifugation. Furthermore, it comprehensively investigates CDsEV contents and cell origins. Small RNA sequencing and proteomics are performed to analyze the miRNAs and proteins of CDsEVs. The miRNAs and proteins of CDsEVs are crucial for maintaining normal auditory function. Among them, FGFR1 in CDsEVs may mediate the survival of cochlear hair cells via sEVs. Finally, the joint analysis of single CDsEV sequencing and single-cell RNA sequencing data is utilized to trace cellular origins of CDsEVs. The results show that different types of cochlear cells secrete different amounts of CDsEVs, with Kölliker's organ cells and supporting cells secrete the most. The findings are expected to enhance the understanding of CDsEVs in the cochlea.
Collapse
Affiliation(s)
- Pei Jiang
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
- Southeast University Shenzhen Research Institute, Shenzhen, 1518063, China
| | - Xiangyu Ma
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
| | - Xinlin Wang
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
| | - Jingyuan Huang
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
| | - Yintao Wang
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
| | - Jingru Ai
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
- Southeast University Shenzhen Research Institute, Shenzhen, 1518063, China
| | - Hairong Xiao
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
- Southeast University Shenzhen Research Institute, Shenzhen, 1518063, China
| | - Mingchen Dai
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
| | - Yanqin Lin
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
- Southeast University Shenzhen Research Institute, Shenzhen, 1518063, China
| | - Buwei Shao
- School of Medicine, Faculty of Medical & Health Sciences, Tel Aviv University, Tel Aviv, 6997801, Israel
| | - Xujun Tang
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
| | - Wei Tong
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
| | - Zixuan Ye
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
- Southeast University Shenzhen Research Institute, Shenzhen, 1518063, China
| | - Renjie Chai
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
- Department of Otolaryngology Head and Neck Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China
- Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Science, Beijing, 100101, China
| | - Shasha Zhang
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
| |
Collapse
|
33
|
Wang K, Lu J, Song C, Qiao M, Li Y, Chang M, Bao H, Qiu Y, Qian B. Extracellular Vesicles Derived from Ligament Tissue Transport Interleukin-17A to Mediate Ligament-To-Bone Crosstalk in Ankylosing Spondylitis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2406876. [PMID: 39308181 PMCID: PMC11633500 DOI: 10.1002/advs.202406876] [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: 06/20/2024] [Revised: 09/09/2024] [Indexed: 10/12/2024]
Abstract
Pathological new bone formation is a critical feature of the progression of ankylosing spondylitis (AS), and spine ankylosis is a distinctive feature of this condition. Ligaments are the primary regions of pathological new bone formation in AS. Here, it is demonstrated that ligament tissue-derived extracellular vesicles (EVs) and their interleukin-17A (IL-17A) cargo mediate the communication between the tissue and other cells. The investigation revealed that IL-17A in EVs can activate the JAK-STAT3 pathway, thereby stimulating the expression of MMP14 in AS ligament. Overexpression of MMP14 can lead to changes in the cytoskeleton and mechanical signaling of mesenchymal stem cells and other cells. These alterations in cellular cytoskeleton and mechanical signaling at ligament sites in patients with AS or in stem cells treated with EVs can result in pathological new bone formation. Finally, inhibiting IL-17A activity and EV endocytosis effectively controlled inflammation and pathological new bone formation. Overall, these data suggest that ligament-derived EVs and the enclosed IL-17A have a potential role in driving pathological new bone formation in AS, and targeting EVs may therefore emerge as a novel approach to delaying ectopic ossification in AS.
Collapse
Affiliation(s)
- Kaiyang Wang
- Division of Spine SurgeryDepartment of Orthopedic SurgeryNanjing Drum Tower HospitalAffiliated Hospital of Medical SchoolNanjing UniversityZhongshan Road 321Nanjing210008China
| | - Jingshun Lu
- Division of Spine SurgeryDepartment of Orthopedic SurgeryNanjing Drum Tower HospitalAffiliated Hospital of Medical SchoolNanjing UniversityZhongshan Road 321Nanjing210008China
| | - Chenyu Song
- Division of Spine SurgeryDepartment of Orthopedic SurgeryNanjing Drum Tower HospitalAffiliated Hospital of Medical SchoolNanjing UniversityZhongshan Road 321Nanjing210008China
| | - Mu Qiao
- Division of Spine SurgeryDepartment of Orthopedic SurgeryNanjing Drum Tower HospitalAffiliated Hospital of Medical SchoolNanjing UniversityZhongshan Road 321Nanjing210008China
| | - Yao Li
- Division of Spine SurgeryDepartment of Orthopedic SurgeryNanjing Drum Tower HospitalAffiliated Hospital of Medical SchoolNanjing UniversityZhongshan Road 321Nanjing210008China
| | - Menghan Chang
- Division of Spine SurgeryDepartment of Orthopedic SurgeryNanjing Drum Tower HospitalAffiliated Hospital of Medical SchoolNanjing UniversityZhongshan Road 321Nanjing210008China
| | - Hongda Bao
- Division of Spine SurgeryDepartment of Orthopedic SurgeryNanjing Drum Tower HospitalAffiliated Hospital of Medical SchoolNanjing UniversityZhongshan Road 321Nanjing210008China
| | - Yong Qiu
- Division of Spine SurgeryDepartment of Orthopedic SurgeryNanjing Drum Tower HospitalAffiliated Hospital of Medical SchoolNanjing UniversityZhongshan Road 321Nanjing210008China
| | - Bang‐Ping Qian
- Division of Spine SurgeryDepartment of Orthopedic SurgeryNanjing Drum Tower HospitalAffiliated Hospital of Medical SchoolNanjing UniversityZhongshan Road 321Nanjing210008China
| |
Collapse
|
34
|
Yang M, Zhou J, Lu L, Deng D, Huang J, Tang Z, Shi X, Lo P, Lovell JF, Zheng Y, Jin H. Tumor cell membrane-based vaccines: A potential boost for cancer immunotherapy. EXPLORATION (BEIJING, CHINA) 2024; 4:20230171. [PMID: 39713208 PMCID: PMC11655317 DOI: 10.1002/exp.20230171] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 03/08/2024] [Indexed: 12/24/2024]
Abstract
Because therapeutic cancer vaccines can, in theory, eliminate tumor cells specifically with relatively low toxicity, they have long been considered for application in repressing cancer progression. Traditional cancer vaccines containing a single or a few discrete tumor epitopes have failed in the clinic, possibly due to challenges in epitope selection, target downregulation, cancer cell heterogeneity, tumor microenvironment immunosuppression, or a lack of vaccine immunogenicity. Whole cancer cell or cancer membrane vaccines, which provide a rich source of antigens, are emerging as viable alternatives. Autologous and allogenic cellular cancer vaccines have been evaluated as clinical treatments. Tumor cell membranes (TCMs) are an intriguing antigen source, as they provide membrane-accessible targets and, at the same time, serve as integrated carriers of vaccine adjuvants and other therapeutic agents. This review provides a summary of the properties and technologies for TCM cancer vaccines. Characteristics, categories, mechanisms, and preparation methods are discussed, as are the demonstrable additional benefits derived from combining TCM vaccines with chemotherapy, sonodynamic therapy, phototherapy, and oncolytic viruses. Further research in chemistry, biomedicine, cancer immunology, and bioinformatics to address current drawbacks could facilitate the clinical adoption of TCM vaccines.
Collapse
Affiliation(s)
- Muyang Yang
- College of Biomedicine and Health and College of Life Science and TechnologyHuazhong Agricultural UniversityWuhanChina
| | - Jie Zhou
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory HealthThe First Affiliated Hospital of Guangzhou Medical UniversityGuangzhouChina
| | - Liseng Lu
- College of Biomedicine and Health and College of Life Science and TechnologyHuazhong Agricultural UniversityWuhanChina
| | - Deqiang Deng
- College of Biomedicine and Health and College of Life Science and TechnologyHuazhong Agricultural UniversityWuhanChina
| | - Jing Huang
- College of Biomedicine and Health and College of Life Science and TechnologyHuazhong Agricultural UniversityWuhanChina
| | - Zijian Tang
- College of Biomedicine and Health and College of Life Science and TechnologyHuazhong Agricultural UniversityWuhanChina
| | - Xiujuan Shi
- College of Biomedicine and Health and College of Life Science and TechnologyHuazhong Agricultural UniversityWuhanChina
| | - Pui‐Chi Lo
- Department of Biomedical SciencesCity University of Hong KongKowloonHong KongChina
| | - Jonathan F. Lovell
- Department of Biomedical EngineeringUniversity at BuffaloState University of New YorkBuffaloNew YorkUSA
| | - Yongfa Zheng
- Department of OncologyRenmin Hospital of Wuhan UniversityWuhanChina
| | - Honglin Jin
- College of Biomedicine and Health and College of Life Science and TechnologyHuazhong Agricultural UniversityWuhanChina
| |
Collapse
|
35
|
Sun X, Setrerrahmane S, Li C, Hu J, Xu H. Nucleic acid drugs: recent progress and future perspectives. Signal Transduct Target Ther 2024; 9:316. [PMID: 39609384 PMCID: PMC11604671 DOI: 10.1038/s41392-024-02035-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 09/20/2024] [Accepted: 10/25/2024] [Indexed: 11/30/2024] Open
Abstract
High efficacy, selectivity and cellular targeting of therapeutic agents has been an active area of investigation for decades. Currently, most clinically approved therapeutics are small molecules or protein/antibody biologics. Targeted action of small molecule drugs remains a challenge in medicine. In addition, many diseases are considered 'undruggable' using standard biomacromolecules. Many of these challenges however, can be addressed using nucleic therapeutics. Nucleic acid drugs (NADs) are a new generation of gene-editing modalities characterized by their high efficiency and rapid development, which have become an active research topic in new drug development field. However, many factors, including their low stability, short half-life, high immunogenicity, tissue targeting, cellular uptake, and endosomal escape, hamper the delivery and clinical application of NADs. Scientists have used chemical modification techniques to improve the physicochemical properties of NADs. In contrast, modified NADs typically require carriers to enter target cells and reach specific intracellular locations. Multiple delivery approaches have been developed to effectively improve intracellular delivery and the in vivo bioavailability of NADs. Several NADs have entered the clinical trial recently, and some have been approved for therapeutic use in different fields. This review summarizes NADs development and evolution and introduces NADs classifications and general delivery strategies, highlighting their success in clinical applications. Additionally, this review discusses the limitations and potential future applications of NADs as gene therapy candidates.
Collapse
Affiliation(s)
- Xiaoyi Sun
- Jiangsu Province Engineering Research Center of Synthetic Peptide Drug Discovery and Evaluation, China Pharmaceutical University, Nanjing, 210009, China
| | | | - Chencheng Li
- Jiangsu Province Engineering Research Center of Synthetic Peptide Drug Discovery and Evaluation, China Pharmaceutical University, Nanjing, 210009, China
| | - Jialiang Hu
- Jiangsu Province Engineering Research Center of Synthetic Peptide Drug Discovery and Evaluation, China Pharmaceutical University, Nanjing, 210009, China
| | - Hanmei Xu
- Jiangsu Province Engineering Research Center of Synthetic Peptide Drug Discovery and Evaluation, China Pharmaceutical University, Nanjing, 210009, China.
| |
Collapse
|
36
|
Li J, Song J, Jia L, Wang M, Ji X, Meng R, Zhou D. Exosomes in Central Nervous System Diseases: A Comprehensive Review of Emerging Research and Clinical Frontiers. Biomolecules 2024; 14:1519. [PMID: 39766226 PMCID: PMC11673277 DOI: 10.3390/biom14121519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 11/20/2024] [Accepted: 11/25/2024] [Indexed: 01/11/2025] Open
Abstract
Exosomes, nano-sized lipid bilayer vesicles, have garnered significant attention as mediators of cell communication, particularly within the central nervous system (CNS). Their unique properties, including high stability, low immunogenicity, and the ability to traverse the blood-brain barrier (BBB), position them as promising tools for understanding and addressing CNS diseases. This comprehensive review delves into the biogenesis, properties, composition, functions, and isolation of exosomes, with a particular focus on their roles in cerebrovascular diseases, neurodegenerative disorders, and CNS tumors. Exosomes are involved in key pathophysiological processes in the CNS, including angiogenesis, inflammation, apoptosis, and cellular microenvironment modification. They demonstrate promise in mitigating ischemic injury, regulating inflammatory responses, and providing neuroprotection across various CNS conditions. Furthermore, exosomes carry distinct biomolecules, offering a novel method for the early diagnosis and monitoring of CNS diseases. Despite their potential, challenges such as complex extraction processes, the heterogeneity of exosomal contents, and targeted delivery limitations hinder their clinical application. Nevertheless, exosomes hold significant promise for advancing our understanding of CNS diseases and developing novel therapeutic strategies. This manuscript significantly contributes to the field by highlighting exosomes' potential in advancing our understanding of CNS diseases, underscoring their unique value in developing novel therapeutic strategies and mediating cellular communication.
Collapse
Affiliation(s)
- Jingrun Li
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
- Advanced Center of Stroke, Beijing Institute for Brain Disorders, Beijing 100053, China
- National Center for Neurological Disorders, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Jiahao Song
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
- Advanced Center of Stroke, Beijing Institute for Brain Disorders, Beijing 100053, China
- National Center for Neurological Disorders, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Lina Jia
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
- Advanced Center of Stroke, Beijing Institute for Brain Disorders, Beijing 100053, China
- National Center for Neurological Disorders, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Mengqi Wang
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
- Advanced Center of Stroke, Beijing Institute for Brain Disorders, Beijing 100053, China
- National Center for Neurological Disorders, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Xunming Ji
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
- Advanced Center of Stroke, Beijing Institute for Brain Disorders, Beijing 100053, China
- National Center for Neurological Disorders, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Ran Meng
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
- Advanced Center of Stroke, Beijing Institute for Brain Disorders, Beijing 100053, China
- National Center for Neurological Disorders, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Da Zhou
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
- Advanced Center of Stroke, Beijing Institute for Brain Disorders, Beijing 100053, China
- National Center for Neurological Disorders, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| |
Collapse
|
37
|
Kim S, Kang Y, Shin H, Lee EB, Ham BJ, Choi Y. Liquid Biopsy-Based Detection and Response Prediction for Depression. ACS NANO 2024; 18:32498-32507. [PMID: 39501510 PMCID: PMC11604100 DOI: 10.1021/acsnano.4c08233] [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: 06/20/2024] [Revised: 10/25/2024] [Accepted: 10/29/2024] [Indexed: 11/27/2024]
Abstract
Proactively predicting antidepressant treatment response before medication failures is crucial, as it reduces unsuccessful attempts and facilitates the development of personalized therapeutic strategies, ultimately enhancing treatment efficacy. The current decision-making process, which heavily depends on subjective indicators, underscores the need for an objective, indicator-based approach. This study developed a method for detecting depression and predicting treatment response through deep learning-based spectroscopic analysis of extracellular vesicles (EVs) from plasma. EVs were isolated from the plasma of both nondepressed and depressed groups, followed by Raman signal acquisition, which was used for AI algorithm development. The algorithm successfully distinguished depression patients from healthy individuals and those with panic disorder, achieving an AUC accuracy of 0.95. This demonstrates the model's capability to selectively diagnose depression within a nondepressed group, including those with other mental health disorders. Furthermore, the algorithm identified depression-diagnosed patients likely to respond to antidepressants, classifying responders and nonresponders with an AUC accuracy of 0.91. To establish a diagnostic foundation, the algorithm applied explainable AI (XAI), enabling personalized medicine for companion diagnostics and highlighting its potential for the development of liquid biopsy-based mental disorder diagnosis.
Collapse
Affiliation(s)
- Seungmin Kim
- Department
of Biomedical Engineering, Korea University, Seoul 02841, Republic of Korea
- Interdisciplinary
Program in Precision Public Health, Korea
University, Seoul 02841, Republic of Korea
| | - Youbin Kang
- Department
of Biomedical Sciences, Korea University
College of Medicine, Seoul 02841, Republic
of Korea
| | - Hyunku Shin
- Exopert
Corporation, Seoul 02841, Republic of Korea
| | - Eun Byul Lee
- Exopert
Corporation, Seoul 02841, Republic of Korea
| | - Byung-Joo Ham
- Department
of Biomedical Sciences, Korea University
College of Medicine, Seoul 02841, Republic
of Korea
| | - Yeonho Choi
- Department
of Biomedical Engineering, Korea University, Seoul 02841, Republic of Korea
- Interdisciplinary
Program in Precision Public Health, Korea
University, Seoul 02841, Republic of Korea
- Exopert
Corporation, Seoul 02841, Republic of Korea
- School
of Bioengineering, Korea University, Seoul 02841, Republic of Korea
| |
Collapse
|
38
|
Romero-Castillo I, López-García A, Diebold Y, García-Posadas L. Enrichment protocols for human conjunctival extracellular vesicles and their characterization. Sci Rep 2024; 14:28270. [PMID: 39550477 PMCID: PMC11569262 DOI: 10.1038/s41598-024-79481-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 11/11/2024] [Indexed: 11/18/2024] Open
Abstract
The understanding of the role played by extracellular vesicles (EVs) in different tissues has improved significantly in the last years, but remains limited concerning the conjunctiva, a complex eye tissue whose role is pivotal for corneal protection. Here, we conducted a comparative study to isolate and characterize EVs from human conjunctival epithelial (IM-HConEpiC) and human conjunctival mesenchymal stromal cell (Conj-MSCs) secretomes using different isolation methods: differential ultracentrifugation (UC), and a combination of ultrafiltration (UF) with precipitation or size exclusion chromatography (SEC). EVs were characterized by total protein content, size, morphology, and expression of protein markers. EV functional effect was tested in an in vitro oxidative stress model. We successfully recovered EVs with the three methods, although significantly higher yields were obtained with UF-precipitation. Dynamic light scattering analysis confirmed the presence of nano-sized particles, being UC-isolated EVs larger than those isolated by UF-precipitation and UF-SEC. Atomic Force Microscopy showed EVs with a slightly ellipsoidal morphology. EVs enriched with UF-precipitation method were further analyzed, confirming the expression of Alix, CD63, TSG101, and Syntenin-1 by Western blotting and showing that Conj-MSC-derived EVs significantly reduced oxidative stress on IM-HConEpiC. Therefore, we conclude that UF-precipitation is the most efficient method for conjunctival EV enrichment.
Collapse
Affiliation(s)
- Ismael Romero-Castillo
- Ocular Surface Group, Instituto Universitario de Oftalmobiología Aplicada (IOBA), Universidad de Valladolid, Valladolid, Spain.
| | - Antonio López-García
- Ocular Surface Group, Instituto Universitario de Oftalmobiología Aplicada (IOBA), Universidad de Valladolid, Valladolid, Spain
| | - Yolanda Diebold
- Ocular Surface Group, Instituto Universitario de Oftalmobiología Aplicada (IOBA), Universidad de Valladolid, Valladolid, Spain
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Laura García-Posadas
- Ocular Surface Group, Instituto Universitario de Oftalmobiología Aplicada (IOBA), Universidad de Valladolid, Valladolid, Spain.
| |
Collapse
|
39
|
Xu Z, Foster JB, Lashley R, Wang X, Muhleman AJ, Masters CE, Lin CLG. Comparison of the protein composition of isolated extracellular vesicles from mouse brain and dissociated brain cell culture medium. PLoS One 2024; 19:e0309716. [PMID: 39531446 PMCID: PMC11556680 DOI: 10.1371/journal.pone.0309716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 08/18/2024] [Indexed: 11/16/2024] Open
Abstract
Extracellular vesicles (EVs) play a crucial role in intercellular communication. Characterizing EV protein composition is essential to understand EV function(s). Isolating EVs from cell culture medium is a common approach to study EVs, but it remains unclear whether EVs isolated from in vitro conditions accurately reflect physiological conditions of the same source in vivo tissues. Here, we analyzed the protein composition of EVs isolated from freshly dissected mouse forebrain and primary dissociated mouse forebrain culture medium. In total, 3,204 and 3,583 proteins were identified in EVs isolated in vivo and in vitro, respectively. Among the proteins identified from both EV sources, there was substantial overlap (~86%). While the overall proteome compositions were very similar, in vitro EVs were relatively enriched with transmembrane/GPI-anchored membrane and cytosolic proteins (MISEV2023 category 1 and 2) typically associated with EVs. Conversely, while both in vivo and in vitro EVs express likely non-EV proteins (MISEV2023 category 3), the in vivo samples were significantly more enriched with these probable contaminants, specifically ribosomal proteins. Our findings highlight that in vitro EVs may be representative of in vivo EVs when isolated from the same source tissue using similar methodology; however, each population of EVs have differences in both total and, primarily, relative protein expression likely due to differing levels of co-eluting contaminants. Therefore, these points must be considered when interpreting results of EV studies further suggesting that improved methods of isolation to reduce non-EV contaminants should be further investigated.
Collapse
Affiliation(s)
- Zan Xu
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, Ohio, United States of America
| | - Joshua Brian Foster
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, Ohio, United States of America
| | - Rashelle Lashley
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, Ohio, United States of America
| | - Xueqin Wang
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, Ohio, United States of America
| | - Albert John Muhleman
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, Ohio, United States of America
| | - Christopher Eli Masters
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, Ohio, United States of America
| | - Chien-liang Glenn Lin
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, Ohio, United States of America
| |
Collapse
|
40
|
Zhang Y, Song M, Fan J, Guo X, Tao S. Impact of probiotics-derived extracellular vesicles on livestock gut barrier function. J Anim Sci Biotechnol 2024; 15:149. [PMID: 39506860 PMCID: PMC11542448 DOI: 10.1186/s40104-024-01102-8] [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: 06/03/2024] [Accepted: 09/05/2024] [Indexed: 11/08/2024] Open
Abstract
Probiotic extracellular vesicles (pEVs) are biologically active nanoparticle structures that can regulate the intestinal tract through direct or indirect mechanisms. They enhance the intestinal barrier function in livestock and poultry and help alleviate intestinal diseases. The specific effects of pEVs depend on their internal functional components, including nucleic acids, proteins, lipids, and other substances. This paper presents a narrative review of the impact of pEVs on the intestinal barrier across various segments of the intestinal tract, exploring their mechanisms of action while highlighting the limitations of current research. Investigating the mechanisms through which probiotics operate via pEVs could deepen our understanding and provide a theoretical foundation for their application in livestock production.
Collapse
Affiliation(s)
- Yuhan Zhang
- College of Animal Sciences and Technology, Huazhong Agricultural University, No. 1 Shizishan Street, Hongshan District, Wuhan, Hubei Province, 430070, China
| | - Mengzhen Song
- College of Animal Sciences and Technology, Huazhong Agricultural University, No. 1 Shizishan Street, Hongshan District, Wuhan, Hubei Province, 430070, China
| | - Jinping Fan
- College of Animal Sciences and Technology, Huazhong Agricultural University, No. 1 Shizishan Street, Hongshan District, Wuhan, Hubei Province, 430070, China
| | - Xuming Guo
- College of Animal Sciences and Technology, Huazhong Agricultural University, No. 1 Shizishan Street, Hongshan District, Wuhan, Hubei Province, 430070, China
| | - Shiyu Tao
- College of Animal Sciences and Technology, Huazhong Agricultural University, No. 1 Shizishan Street, Hongshan District, Wuhan, Hubei Province, 430070, China.
| |
Collapse
|
41
|
Xu F, Chen M, Lin Y, Zhou S, Li J, Yu Y, Xu J, Wu W, Chen Y, Zhang H, Wei Y, Wang W. Functional Three-Dimensional Zeolitic Imidazolate Framework with an Ordered Macroporous Structure for the Isolation of Extracellular Vesicles. Anal Chem 2024; 96:17640-17648. [PMID: 39440634 DOI: 10.1021/acs.analchem.4c03566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2024]
Abstract
Extracellular vesicles (EVs) and their cargoes are increasingly being recognized as noninvasive diagnostic markers, necessitating the isolation of EVs from complex biological samples. Herein, a distearoyl phospholipid ethanolamine-functionalized single-crystal ordered macroporous three-dimensional zeolitic imidazolate framework (SOM-ZIF-8-DSPE) was developed, which combines the surface charge interaction of ZIF-8 with the synergistic effect of DSPE insertion into the phospholipid membrane of EVs to improve the isolating selectivity of EV capture. The materials have porous structures larger than 300 nm in diameter, providing enough space and active sites to trap EVs. Benefiting from this feature, the entire isolation process takes only 10 min and is well compatible with the subsequent analysis of RNA in EVs. Consequently, 10 upregulated miRNA of plasma EVs in the primary colorectal cancer (pCRC) patients is found over the healthy donors, and 6 upregulated miRNA of plasma EVs in the metastatic colorectal cancer (mCRC) patients over pCRC patients. These findings suggest that the isolation of EV-based SOM-ZIF-8-DSPE is a promising strategy to identify biomarkers for disease diagnosis, such as miRNAs in plasma EVs for the early detection of CRC.
Collapse
Affiliation(s)
- Fang Xu
- College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Mengxi Chen
- College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Yujie Lin
- College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Shenyue Zhou
- College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Jiaxi Li
- College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Yuanyuan Yu
- College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Jiayu Xu
- College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Wen Wu
- College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Yinshuang Chen
- College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Haiyang Zhang
- College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Yinmao Wei
- Key Laboratory of Synthetic and Natural Functional Molecule Chemistry of Ministry of Education, College of Chemistry and Materials Science, Northwest University, Xi'an 710127, China
| | - Weipeng Wang
- College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| |
Collapse
|
42
|
Rinaldi A, Balietti M, Principi E, De Luca M, De Felice E, Narcisi FM, Vilardo L, Rosito M, Piacentini R, D'Alessandro G, D'Agnano I, Maggi L, Conti F, Limatola C, Catalano M. BV2-derived extracellular vesicles modulate microglia inflammatory profile, neuronal plasticity, and behavioural performances in late adult mice. Brain Behav Immun 2024; 122:58-74. [PMID: 39128568 DOI: 10.1016/j.bbi.2024.08.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 07/24/2024] [Accepted: 08/08/2024] [Indexed: 08/13/2024] Open
Abstract
BACKGROUND During aging, both the brain and the immune system undergo a progressive impairment of physiological functions. Microglia, the immunocompetent cells of the central nervous system, shift towards a chronic mild inflammatory state that impacts brain homeostasis. Extracellular vesicles (EVs) released by microglia transport packages of molecular information that mirror the inflammatory status of donor cells and modulate the inflammatory phenotype of recipient microglia and other cell types. RESULTS We demonstrated that intranasal administration of EVs derived from microglial-like BV2 cells to late adult mice (16-20 months of age) shifts microglia toward a "juvenile" morphology affecting their inflammatory profile. Mice treated with BV2-derived EVs have a reduction of anxiety-like behavior and an increased spatial learning, with sex-dependent differences. Further, BV2-derived EVs increased neuronal plasticity both in male and female mice. These findings suggest the involvement of microglial cells in vesicles-mediated anti-aging effect. CONCLUSIONS Our data indicate that BV2-derived EVs could represent a resource to slow down age-dependent inflammation in the mouse brain.
Collapse
Affiliation(s)
- Arianna Rinaldi
- Department of Physiology and Pharmacology, Sapienza University, Rome, Italy
| | - Marta Balietti
- IRCCS INRCA, Center for Neurobiology of Aging, Via Birarelli 8, Ancona 60121, Italy
| | - Elisa Principi
- Università Politecnica delle Marche, Department of Experimental and Clinical Medicine, Via Tronto 10/a, Ancona 60126, Italy
| | | | - Eleonora De Felice
- Department of Physiology and Pharmacology, Sapienza University, Rome, Italy
| | | | - Laura Vilardo
- Institute of Biomedical Technologies, CNR, 20054 Segrate, Italy
| | - Maria Rosito
- Department of Physiology and Pharmacology, Sapienza University, Rome, Italy; Center for Life Nanoscience & Neuroscience Istituto Italiano di Tecnologia@Sapienza, Rome, Italy
| | - Roberto Piacentini
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy; IRCCS Fondazione Policlinico Universitario A. Gemelli, Largo A. Gemelli 1, Roma, Italy
| | - Giuseppina D'Alessandro
- Department of Physiology and Pharmacology, Sapienza University, Rome, Italy; IRCCS Neuromed, Pozzilli, IS, Italy
| | - Igea D'Agnano
- Institute of Biomedical Technologies, CNR, 20054 Segrate, Italy
| | - Laura Maggi
- Department of Physiology and Pharmacology, Sapienza University, Rome, Italy
| | - Fiorenzo Conti
- Università Politecnica delle Marche, Department of Experimental and Clinical Medicine, Via Tronto 10/a, Ancona 60126, Italy; IRCCS INRCA, Center for Neurobiology of Aging, Via Birarelli 8, Ancona 60121, Italy
| | - Cristina Limatola
- IRCCS Neuromed, Pozzilli, IS, Italy; Department of Physiology and Pharmacology, Sapienza University, Laboratory affiliated to Institute Pasteur Italia, Rome, Italy
| | - Myriam Catalano
- Department of Physiology and Pharmacology, Sapienza University, Rome, Italy.
| |
Collapse
|
43
|
Matamoros‐Angles A, Karadjuzovic E, Mohammadi B, Song F, Brenna S, Meister SC, Siebels B, Voß H, Seuring C, Ferrer I, Schlüter H, Kneussel M, Altmeppen HC, Schweizer M, Puig B, Shafiq M, Glatzel M. Efficient enzyme-free isolation of brain-derived extracellular vesicles. J Extracell Vesicles 2024; 13:e70011. [PMID: 39508423 PMCID: PMC11541858 DOI: 10.1002/jev2.70011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 10/09/2024] [Accepted: 10/16/2024] [Indexed: 11/15/2024] Open
Abstract
Extracellular vesicles (EVs) have gained significant attention as pathology mediators and potential diagnostic tools for neurodegenerative diseases. However, isolation of brain-derived EVs (BDEVs) from tissue remains challenging, often involving enzymatic digestion steps that may compromise the integrity of EV proteins and overall functionality. Here, we describe that collagenase digestion, commonly used for BDEV isolation, produces undesired protein cleavage of EV-associated proteins in brain tissue homogenates and cell-derived EVs. In order to avoid this effect, we studied the possibility of isolating BDEVs with a reduced amount of collagenase or without any protease. Characterization of the isolated BDEVs from mouse and human samples (both female and male) revealed their characteristic morphology and size distribution with both approaches. However, we show that even minor enzymatic digestion induces 'artificial' proteolytic processing in key BDEV markers, such as Flotillin-1, CD81, and the cellular prion protein (PrPC), whereas avoiding enzymatic treatment completely preserves their integrity. We found no major differences in mRNA and protein content between non-enzymatically and enzymatically isolated BDEVs, suggesting that the same BDEV populations are purified with both approaches. Intriguingly, the lack of Golgi marker GM130 signal, often referred to as contamination indicator (or negative marker) in EV preparations, seems to result from enzymatic digestion rather than from its actual absence in BDEV samples. Overall, we show that non-enzymatic isolation of EVs from brain tissue is possible and avoids artificial pruning of proteins while achieving an overall high BDEV yield and purity. This protocol will help to understand the functions of BDEV and their associated proteins in a near-physiological setting, thus opening new research approaches.
Collapse
Affiliation(s)
| | - Emina Karadjuzovic
- Institute of NeuropathologyUniversity Medical Center Hamburg‐Eppendorf (UKE)HamburgGermany
| | - Behnam Mohammadi
- Institute of NeuropathologyUniversity Medical Center Hamburg‐Eppendorf (UKE)HamburgGermany
| | - Feizhi Song
- Institute of NeuropathologyUniversity Medical Center Hamburg‐Eppendorf (UKE)HamburgGermany
| | - Santra Brenna
- Department of Neurology, Experimental Research in Stroke and Inflammation (ERSI)University Medical Center Hamburg‐Eppendorf (UKE)HamburgGermany
| | | | - Bente Siebels
- Section Mass Spectrometry and ProteomicsUniversity Medical Center Hamburg‐Eppendorf (UKE)HamburgGermany
| | - Hannah Voß
- Section Mass Spectrometry and ProteomicsUniversity Medical Center Hamburg‐Eppendorf (UKE)HamburgGermany
| | - Carolin Seuring
- Multi‐User‐CryoEM‐FacilityCentre for Structural Systems Biology (CSSB)HamburgGermany
- Department of ChemistryUniversität HamburgHamburgGermany
- Leibniz Institute of Virology (LIV)HamburgGermany
| | - Isidre Ferrer
- IDIBELLUniversity of BarcelonaL'Hospitalet de LlobregatSpain
| | - Hartmut Schlüter
- Section Mass Spectrometry and ProteomicsUniversity Medical Center Hamburg‐Eppendorf (UKE)HamburgGermany
| | - Matthias Kneussel
- Institute for Molecular Neurogenetics, Center for Molecular Neurobiology Hamburg (ZMNH)University Medical Center Hamburg‐Eppendorf (UKE)HamburgGermany
| | | | - Michaela Schweizer
- Electron Microscopy Core Facility, Center for Molecular Neurobiology (ZMNH)University Medical Center Hamburg‐Eppendorf (UKE)HamburgGermany
| | - Berta Puig
- Department of Neurology, Experimental Research in Stroke and Inflammation (ERSI)University Medical Center Hamburg‐Eppendorf (UKE)HamburgGermany
| | - Mohsin Shafiq
- Institute of NeuropathologyUniversity Medical Center Hamburg‐Eppendorf (UKE)HamburgGermany
| | - Markus Glatzel
- Institute of NeuropathologyUniversity Medical Center Hamburg‐Eppendorf (UKE)HamburgGermany
| |
Collapse
|
44
|
Wang H, Wang Y, Wang S, Ling M, Luo J, Sun J, Xi Q, Chen T, Zhang Y. Assessment of isolation strategies to remove caseins for high-quality milk-derived extracellular vesicles. J Dairy Sci 2024; 107:8934-8946. [PMID: 39033914 DOI: 10.3168/jds.2024-25162] [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: 05/13/2024] [Accepted: 06/27/2024] [Indexed: 07/23/2024]
Abstract
Increasing studies have highlighted the significance of milk-derived extracellular vesicles (MEV) in mother-newborn integration, as well as their application as novel drug delivery systems and diagnostic biomarkers. However, conventional ultracentrifugation (UC) often results in the co-precipitation of casein micelles in MEV pellets. In this study, we compared methods with different principles to screen the optimal pretreatment in caseins removal, and found that isoelectric precipitation by hydrochloric acid (HA) could most effectively remove caseins in porcine milk. We further characterized MEV populations isolated by UC and HA/UC from diverse aspects, including particle methodology via nanoparticle tracking analysis (NTA) and transmission electron microscopy (TEM), RNA and protein contents, and purity analysis. Importantly, the proliferative and anti-inflammatory effects of MEV were evaluated in vitro, showing the superiority of MEV via HA/UC in functionality compared with UC. Our results suggest that HA pretreatment before ultracentrifugation could effectively remove caseins and other protein complexes, making MEV with higher purity and more significant effects in vitro. This study provides valuable insights for the advancement of MEV isolation techniques across different species and accurate function analysis of MEV.
Collapse
Affiliation(s)
- Hailong Wang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Guangdong Laboratory for Lingnan Modern Agriculture, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Yuxuan Wang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Guangdong Laboratory for Lingnan Modern Agriculture, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Shumeng Wang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Guangdong Laboratory for Lingnan Modern Agriculture, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Mingwang Ling
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Guangdong Laboratory for Lingnan Modern Agriculture, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Junyi Luo
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Guangdong Laboratory for Lingnan Modern Agriculture, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Jiajie Sun
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Guangdong Laboratory for Lingnan Modern Agriculture, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Qianyun Xi
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Guangdong Laboratory for Lingnan Modern Agriculture, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Ting Chen
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Guangdong Laboratory for Lingnan Modern Agriculture, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Yongliang Zhang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Guangdong Laboratory for Lingnan Modern Agriculture, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China.
| |
Collapse
|
45
|
Moreira RS, Benetti Filho V, Maia GA, Soratto TAT, Kawagoe EK, Russi BC, Miletti LC, Wagner G. FastProtein-an automated software for in silico proteomic analysis. PeerJ 2024; 12:e18309. [PMID: 39494269 PMCID: PMC11531748 DOI: 10.7717/peerj.18309] [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: 04/30/2024] [Accepted: 09/24/2024] [Indexed: 11/05/2024] Open
Abstract
Although various tools provide proteomic information, each tool has limitations related to execution platforms, libraries, versions, and data output format. Integrating data generated from different software is a laborious process that can prolong analysis time. Here, we present FastProtein, a protein analysis pipeline that is user-friendly, easily installable, and outputs important information about subcellular location, transmembrane domains, signal peptide, molecular weight, isoelectric point, hydropathy, aromaticity, gene ontology, endoplasmic reticulum retention domains, and N-glycosylation domains. It also helps determine the presence of glycosylphosphatidylinositol and obtain functional information from InterProScan, PANTHER, Pfam, and alignment-based annotation searches. FastProtein provides the scientific community with an easy-to-use computational tool for proteomic data analysis. It is applicable to both small datasets and proteome-wide studies. It can be used through the command line interface mode or a web interface installed on a local server. FastProtein significantly enhances proteomics analysis workflows by producing multiple results in a single-step process, thereby streamlining and accelerating the overall analysis. The software is open-source and freely available. Installation and execution instructions, as well as the source code and test files generated for tool validation, are available at https://github.com/bioinformatics-ufsc/FastProtein.
Collapse
Affiliation(s)
- Renato Simões Moreira
- Instituto Federal de Santa Catarina, Gaspar, Santa Catarina, Brazil
- Departamento de Microbiologia, Parasitologia e Imunologia, Universidade Federal de Santa Catarina, Florianópolis, Santa Catarina, Brazil
| | - Vilmar Benetti Filho
- Departamento de Microbiologia, Parasitologia e Imunologia, Universidade Federal de Santa Catarina, Florianópolis, Santa Catarina, Brazil
| | - Guilherme Augusto Maia
- Departamento de Microbiologia, Parasitologia e Imunologia, Universidade Federal de Santa Catarina, Florianópolis, Santa Catarina, Brazil
| | - Tatiany Aparecida Teixeira Soratto
- Departamento de Microbiologia, Parasitologia e Imunologia, Universidade Federal de Santa Catarina, Florianópolis, Santa Catarina, Brazil
| | - Eric Kazuo Kawagoe
- Departamento de Microbiologia, Parasitologia e Imunologia, Universidade Federal de Santa Catarina, Florianópolis, Santa Catarina, Brazil
| | | | - Luiz Cláudio Miletti
- Centro de Ciências Agroveterinárias, Universidade do Estado de Santa Catarina, Lages, Santa Catarina, Brazil
| | - Glauber Wagner
- Departamento de Microbiologia, Parasitologia e Imunologia, Universidade Federal de Santa Catarina, Florianópolis, Santa Catarina, Brazil
| |
Collapse
|
46
|
Xiao J, Deng Y, Xie J, Liu H, Yang Q, Zhang Y, Huang X, Cao Z. Apoptotic vesicles from macrophages exacerbate periodontal bone resorption in periodontitis via delivering miR-143-3p targeting Igfbp5. J Nanobiotechnology 2024; 22:658. [PMID: 39456001 PMCID: PMC11515254 DOI: 10.1186/s12951-024-02934-2] [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: 07/24/2024] [Accepted: 10/14/2024] [Indexed: 10/28/2024] Open
Abstract
ABSTRCT BACKGROUND: Apoptotic vesicles (ApoVs), which are extracellular vesicles released by apoptotic cells, have been reported to exhibit substantial therapeutic potential for inflammatory diseases and tissue regeneration. While extensive research has been dedicated to mesenchymal stem cells (MSCs), the investigation into immune cell-derived ApoVs remains limited, particularly regarding the function and fate of macrophage-derived ApoVs in the context of periodontitis (PD). RESULTS Our study corroborates the occurrence and contribution of resident macrophage apoptosis in Porphyromonas gingivalis (Pg)-associated PD. The findings unveil the pivotal role played by apoptotic macrophages and their derived ApoVs in orchestrating periodontal bone remodeling. The enrichments of diverse functional miRNAs within these ApoVs are discerned through sequencing techniques. Moreover, our study elucidates that the macrophage-derived ApoVs predominantly deliver miR-143-3p, targeting insulin-like growth factor-binding protein 5 (IGFBP5), thereby disrupting periodontal bone homeostasis. CONCLUSIONS Our study reveals that macrophages in Pg-associated PD undergo apoptosis and generate miR-143-3p-enriched ApoVs to silence IGFBP5, resulting in the perturbation of osteogenic-osteoclastic balance and the ensuing periodontal bone destruction. Accordingly, interventions targeting miR-143-3p in macrophages or employment of apoptosis inhibitor Z-VAD hold promise as effective therapeutic strategies for the management of PD.
Collapse
Affiliation(s)
- Junhong Xiao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
- Qingdao Stomatological Hospital Affiliated to Qingdao University, No.17 Dexian Road, Shinan District, Qingdao, 266001, Shandong Province, China
| | - Yifei Deng
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Jirong Xie
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Heyu Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Qiudong Yang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Yufeng Zhang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China.
- Medical Research Institute, School of Medicine, Wuhan University, Wuhan, 430071, China.
- Department of Oral Implantology, School & Hospital of Stomatology, Wuhan University, 237 Luoyu Road, Hongshan District, Wuhan, 430079, China.
| | - Xin Huang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China.
- Department of Periodontology, School & Hospital of Stomatology, Wuhan University, 237 Luoyu Road, Hongshan District, Wuhan, 430079, China.
| | - Zhengguo Cao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China.
- Department of Periodontology, School & Hospital of Stomatology, Wuhan University, 237 Luoyu Road, Hongshan District, Wuhan, 430079, China.
| |
Collapse
|
47
|
Guo S, Wang X, Shan D, Xiao Y, Ju L, Zhang Y, Wang G, Qian K. The detection, biological function, and liquid biopsy application of extracellular vesicle-associated DNA. Biomark Res 2024; 12:123. [PMID: 39402599 PMCID: PMC11476736 DOI: 10.1186/s40364-024-00661-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 09/24/2024] [Indexed: 10/19/2024] Open
Abstract
Cell-derived extracellular vesicles (EVs), which carry diverse biomolecules such as nucleic acids, proteins, metabolites, and lipids reflecting their cell of origin, are released under both physiological and pathological conditions. EVs have been demonstrated to mediate cell-to-cell communication and serve as biomarkers. EV-associated DNA (EV-DNA) comprises genomic and mitochondrial DNA (i.e., gDNA and mtDNA) fragments. Some studies have revealed that EV-DNA can represent the full nuclear genome and mitochondrial genome of parental cells. Furthermore, DNA fragments loaded into EVs are stable and can be transferred to recipient cells to regulate their biological functions. In this review, we summarized and discussed EV-DNA research advances with an emphasis on EV-DNA detection at the population-EV and single-EV levels, gene transfer-associated biological functions, and clinical applications as biomarkers for disease liquid biopsy. We hope that this review will provide potential directions or guidance for future EV-DNA investigations.
Collapse
Affiliation(s)
- Shan Guo
- Department of Biological Repositories, Human Genetic Resources Preservation Center of Hubei Province, Zhongnan Hospital of Wuhan University, Wuhan, China.
| | - Xin Wang
- Center for Disease Control and Prevention of Hubei Province, Wuhan, China
| | - Danni Shan
- Department of Biological Repositories, Human Genetic Resources Preservation Center of Hubei Province, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yu Xiao
- Department of Biological Repositories, Human Genetic Resources Preservation Center of Hubei Province, Zhongnan Hospital of Wuhan University, Wuhan, China
- Department of Urology, Hubei Key Laboratory of Urological Diseases, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Lingao Ju
- Department of Urology, Hubei Key Laboratory of Urological Diseases, Zhongnan Hospital of Wuhan University, Wuhan, China
- Human Genetic Resources Preservation Center, Wuhan University, Wuhan, China
| | - Yi Zhang
- Euler Technology, ZGC Life Sciences Park, Beijing, China
- Center for Quantitative Biology, School of Life Sciences, Peking University, Beijing, China
| | - Gang Wang
- Department of Urology, Hubei Key Laboratory of Urological Diseases, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Kaiyu Qian
- Department of Biological Repositories, Human Genetic Resources Preservation Center of Hubei Province, Zhongnan Hospital of Wuhan University, Wuhan, China.
- Wuhan Research Center for Infectious Diseases and Cancer, Chinese Academy of Medical Sciences, Wuhan, China.
| |
Collapse
|
48
|
Chen L, Xu YX, Wang YS, Ren YY, Dong XM, Wu P, Xie T, Zhang Q, Zhou JL. Prostate cancer microenvironment: multidimensional regulation of immune cells, vascular system, stromal cells, and microbiota. Mol Cancer 2024; 23:229. [PMID: 39395984 PMCID: PMC11470719 DOI: 10.1186/s12943-024-02137-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 09/23/2024] [Indexed: 10/14/2024] Open
Abstract
BACKGROUND Prostate cancer (PCa) is one of the most prevalent malignancies in males worldwide. Increasing research attention has focused on the PCa microenvironment, which plays a crucial role in tumor progression and therapy resistance. This review aims to provide a comprehensive overview of the key components of the PCa microenvironment, including immune cells, vascular systems, stromal cells, and microbiota, and explore their implications for diagnosis and treatment. METHODS Keywords such as "prostate cancer", "tumor microenvironment", "immune cells", "vascular system", "stromal cells", and "microbiota" were used for literature retrieval through online databases including PubMed and Web of Science. Studies related to the PCa microenvironment were selected, with a particular focus on those discussing the roles of immune cells, vascular systems, stromal cells, and microbiota in the development, progression, and treatment of PCa. The selection criteria prioritized peer-reviewed articles published in the last five years, aiming to summarize and analyze the latest research advancements and clinical relevance regarding the PCa microenvironment. RESULTS The PCa microenvironment is highly complex and dynamic, with immune cells contributing to immunosuppressive conditions, stromal cells promoting tumor growth, and microbiota potentially affecting androgen metabolism. Vascular systems support angiogenesis, which fosters tumor expansion. Understanding these components offers insight into the mechanisms driving PCa progression and opens avenues for novel therapeutic strategies targeting the tumor microenvironment. CONCLUSIONS A deeper understanding of the PCa microenvironment is crucial for advancing diagnostic techniques and developing precision therapies. This review highlights the potential of targeting the microenvironment to improve patient outcomes, emphasizing its significance in the broader context of PCa research and treatment innovation.
Collapse
Affiliation(s)
- Lin Chen
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
| | - Yu-Xin Xu
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
| | - Yuan-Shuo Wang
- School of Pharmacy, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Ying-Ying Ren
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
| | - Xue-Man Dong
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
| | - Pu Wu
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
| | - Tian Xie
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China.
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China.
| | - Qi Zhang
- Department of Urology, Zhejiang Provincial People's Hospital, Hangzhou, Zhejiang, 310014, China.
| | - Jian-Liang Zhou
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China.
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China.
| |
Collapse
|
49
|
Gurriaran-Rodriguez U, De Repentigny Y, Kothary R, Rudnicki MA. Isolation of small extracellular vesicles from regenerating muscle tissue using tangential flow filtration and size exclusion chromatography. Skelet Muscle 2024; 14:22. [PMID: 39394606 PMCID: PMC11468478 DOI: 10.1186/s13395-024-00355-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Accepted: 09/30/2024] [Indexed: 10/13/2024] Open
Abstract
We have recently made the strikingly discovery that upon a muscle injury, Wnt7a is upregulated and secreted from new regenerating myofibers on the surface of exosomes to elicit its myogenerative response distally. Despite recent advances in extracellular vesicle (EVs) isolation from diverse tissues, there is still a lack of specific methodology to purify EVs from muscle tissue. To eliminate contamination with non-EV secreted proteins and cytoplasmic fragments, which are typically found when using classical methodology, such as ultracentrifugation, we adapted a protocol combining Tangential Flow Filtration (TFF) and Size Exclusion Chromatography (SEC). We found that this approach allows simultaneous purification of Wnt7a, bound to EVs (retentate fraction) and free non-EV Wnt7a (permeate fraction). Here we described this optimized protocol designed to specifically isolate EVs from hind limb muscle explants, without cross-contamination with other sources of non-EV bounded proteins. The first step of the protocol is to remove large EVs with sequential centrifugation. Extracellular vesicles are then concentrated and washed in exchange buffer by TFF. Lastly, SEC is performed to remove any soluble protein traces remaining after TFF. Overall, this procedure can be used to isolate EVs from conditioned media or biofluid that contains EVs derived from any cell type or tissue, improving reproducibility, efficiency, and purity of EVs preparations. Our purification protocol results in high purity EVs that maintain structural integrity and thus fully compatible with in vitro and in vivo bioactivity and analytic assays.
Collapse
Affiliation(s)
- Uxia Gurriaran-Rodriguez
- Ottawa Hospital Research Institute, Regenerative Medicine Program, Ottawa, ON, Canada.
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada.
- CIC bioGUNE, Bizkaia Technology Park, Derio, 48160, Spain.
| | - Yves De Repentigny
- Ottawa Hospital Research Institute, Regenerative Medicine Program, Ottawa, ON, Canada
| | - Rashmi Kothary
- Ottawa Hospital Research Institute, Regenerative Medicine Program, Ottawa, ON, Canada
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Michael A Rudnicki
- Ottawa Hospital Research Institute, Regenerative Medicine Program, Ottawa, ON, Canada.
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada.
- Regenerative Medicine Program, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, ON, K1H 8L6, Canada.
| |
Collapse
|
50
|
Chen ZK, Zheng S, Long Y, Wang KM, Xiao BL, Li JB, Zhang W, Song H, Chen G. High-throughput screening identifies ibuprofen as an sEV PD-L1 inhibitor for synergistic cancer immunotherapy. Mol Ther 2024; 32:3580-3596. [PMID: 39217416 PMCID: PMC11489553 DOI: 10.1016/j.ymthe.2024.08.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 07/13/2024] [Accepted: 08/29/2024] [Indexed: 09/04/2024] Open
Abstract
Programmed death-ligand 1 (PD-L1) on tumor-derived small extracellular vesicles (sEVs) limits therapeutic effectiveness by interacting with the PD-1 receptor on host immune cells. Targeting the secretion of sEV PD-L1 has emerged as a promising strategy to enhance immunotherapy. However, the lack of small-molecule inhibitors poses a challenge for clinical translation. In this study, we developed a target and phenotype dual-driven high-throughput screening strategy that combined virtual screening with nanoflow-based experimental verification. We identified ibuprofen (IBP) as a novel inhibitor that effectively targeted sEV PD-L1 secretion. IBP disrupted the biogenesis and secretion of PD-L1+ sEVs in tumor cells by physically interacting with a critical regulator of sEV biogenesis, hepatocyte growth factor-regulated tyrosine kinase substrate. Notably, the mechanism of action of IBP is distinct from its commonly known targets, cyclooxygenases. Administration of IBP stimulated antitumor immunity and enhanced the efficacy of anti-PD-1 therapy in melanoma and oral squamous cell carcinoma mouse models. To address potential adverse effects, we further developed an IBP gel for topical application, which demonstrated remarkable therapeutic efficacy when combined with anti-PD-1 treatment. The discovery of this specific small inhibitor provides a promising avenue for establishing durable, systemic antitumor immunity.
Collapse
Affiliation(s)
- Zhuo-Kun Chen
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Shuo Zheng
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430079, China
| | - Yan Long
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430079, China
| | - Kui-Ming Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Bo-Lin Xiao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Jin-Bang Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Wei Zhang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China; Department of Oral and Maxillofacial Surgery, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China.
| | - Heng Song
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430079, China.
| | - Gang Chen
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China; Department of Oral and Maxillofacial Surgery, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China; TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan 430079, China; Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan 430079, China.
| |
Collapse
|