1
|
Zhang J, Su N, Liu W, Li M, Zheng H, Li B, Jin X, Gao M, Zhang X. An effective cell-penetrating peptide-based loading method to extracellular vesicles and enhancement in cellular delivery of drugs. Anal Bioanal Chem 2025:10.1007/s00216-025-05742-1. [PMID: 39836222 DOI: 10.1007/s00216-025-05742-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 12/22/2024] [Accepted: 01/06/2025] [Indexed: 01/22/2025]
Abstract
Extracellular vesicles (EVs) have been demonstrated to own the advantages in evading phagocytosis, crossing biological barriers, and possessing excellent biocompatibility and intrinsic stability. Based on these characteristics, EVs have been used as effective therapeutic carriers for drug delivery, but the low drug loading capacity greatly limits further applications. Herein, we developed a drug loading method based on cell-penetrating peptide (CPP) to enhance the encapsulation of therapeutic reagents in EVs, and EVs-based drug delivery system achieved higher killing efficacy to tumor cells. Urinary EVs and chemotherapy reagent doxorubicin (DOX) were used as model. It is easy to conjugate CPP with DOX (CPP-DOX) through the linker N-succinimidyl 3-maleimidopropionate (SMP). CPP-DOX was incubated with EVs under a mild condition, promoting the encapsulation of DOX into EV cavities. CPP-DOX-EVs showed strong anticancer ability since EVs delivery facilitated the uptake by cancer cells. EVs loading of CPP-DOX exhibited higher drug loading efficiency at 37.18%, presenting about 2.5 times increase in efficiency over EVs loading of DOX through passive incubation. Easy operation and controllable condition further reinforce the advantages compared with other loading methods. CPP-based drug loading method provides an effective strategy for EVs-based drug delivery system.
Collapse
Affiliation(s)
- Jin Zhang
- Department of Chemistry and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200433, China
| | - Ning Su
- Department of Chemistry and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200433, China
| | - Wei Liu
- Department of Chemistry and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200433, China
| | - Mengran Li
- Department of Chemistry and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200433, China
| | - Haoyang Zheng
- Department of Chemistry and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200433, China
| | - Bing Li
- Department of Chemistry and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200433, China
| | | | - Mingxia Gao
- Department of Chemistry and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200433, China.
| | - Xiangmin Zhang
- Department of Chemistry and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200433, China
| |
Collapse
|
2
|
Huang G, He Y, Chen X, Yin T, Ma A, Zhu L, Chen L, Liang R, Zhang P, Pan H, Cai L. Bioorthogonal oncolytic-virus nanovesicles combined bio-immunotherapy with CAR-T cells for solid tumors. Biomater Sci 2025; 13:457-465. [PMID: 39607022 DOI: 10.1039/d4bm01305k] [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: 11/29/2024]
Abstract
Various oncolytic viruses (OVs) have been adopted as therapeutic tools to increase the efficacy of chimeric antigen receptor (CAR)-T cells against solid tumors. However, the therapeutic effect of OVs has been limited by pre-existing neutralizing antibodies and poor targeting delivery for systemic administration. Herein, we propose using bioorthogonal OV nanovesicles to boost the antitumor effects of CAR-T cells in solid tumors by reshaping the tumor microenvironment. Using a cell-membrane nanomimetic technique, we embedded artificial chemical ligands on cancer cell surfaces and then encapsulated lysoviral particles to obtain dual-targeted OV nanovesicles with bioorthogonal targeting and homologous recognition. OVs can be directly encapsulated into cancer cell nanovesicles and exhibit a liposome-like nanostructure, efficient loading, and excellent tumor-targeting capability. Encouragingly, OV nanovesicles efficiently induced tumor-cell apoptosis while sparing normal tissues and cells, thereby inhibiting tumor growth. Administration of viral nanovesicles effectively increased the secretion of anti-tumor cytokines such as IL-2, TNF-α and IFN-γ, and significantly promoted the infiltration and activation of CD8+CAR-T cells in tumors. Our data suggest that bioorthogonal OV nanovesicles hold great potential to overcome the limitations of CAR-T cells as monotherapies against solid tumors and, thus, drive the clinical application of combination therapy.
Collapse
Affiliation(s)
- Guojun Huang
- Guangdong Key Laboratory of Nanomedicine, CAS-HK Joint Lab of Biomaterials, Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences (CAS), Shenzhen, 518055, China.
| | - Yiran He
- Guangdong Key Laboratory of Nanomedicine, CAS-HK Joint Lab of Biomaterials, Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences (CAS), Shenzhen, 518055, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xiaocong Chen
- Guangdong Key Laboratory of Nanomedicine, CAS-HK Joint Lab of Biomaterials, Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences (CAS), Shenzhen, 518055, China.
| | - Ting Yin
- Guangdong Key Laboratory of Nanomedicine, CAS-HK Joint Lab of Biomaterials, Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences (CAS), Shenzhen, 518055, China.
| | - Aiqing Ma
- Guangdong Key Laboratory of Nanomedicine, CAS-HK Joint Lab of Biomaterials, Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences (CAS), Shenzhen, 518055, China.
| | - Lizhen Zhu
- Guangdong Key Laboratory of Nanomedicine, CAS-HK Joint Lab of Biomaterials, Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences (CAS), Shenzhen, 518055, China.
| | - Liqi Chen
- Guangdong Key Laboratory of Nanomedicine, CAS-HK Joint Lab of Biomaterials, Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences (CAS), Shenzhen, 518055, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Ruijing Liang
- Guangdong Key Laboratory of Nanomedicine, CAS-HK Joint Lab of Biomaterials, Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences (CAS), Shenzhen, 518055, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Pengfei Zhang
- Guangdong Key Laboratory of Nanomedicine, CAS-HK Joint Lab of Biomaterials, Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences (CAS), Shenzhen, 518055, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Hong Pan
- Guangdong Key Laboratory of Nanomedicine, CAS-HK Joint Lab of Biomaterials, Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences (CAS), Shenzhen, 518055, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Lintao Cai
- Guangdong Key Laboratory of Nanomedicine, CAS-HK Joint Lab of Biomaterials, Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences (CAS), Shenzhen, 518055, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China
- Sino-European Center of Biomedicine and Health, Luohu, Shenzhen 518024, China
| |
Collapse
|
3
|
Aalhate M, Mahajan S, Dhuri A, Singh PK. Biohybrid nano-platforms manifesting effective cancer therapy: Fabrication, characterization, challenges and clinical perspective. Adv Colloid Interface Sci 2025; 335:103331. [PMID: 39522420 DOI: 10.1016/j.cis.2024.103331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 10/01/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024]
Abstract
Nanotechnology-based delivery systems have brought a paradigm shift in the management of cancer. However, the main obstacles to nanocarrier-based delivery are their limited circulation duration, excessive immune clearance, inefficiency in interacting effectively in a biological context and overcoming biological barriers. This demands effective engineering of nanocarriers to achieve maximum efficacy. Nanocarriers can be maneuvered with biological components to acquire biological identity for further regulating their biodistribution and cell-to-cell cross-talk. Thus, the integration of synthetic and biological components to deliver therapeutic cargo is called a biohybrid delivery system. These delivery systems possess the advantage of synthetic nanocarriers, such as high drug loading, engineerable surface, reproducibility, adequate communication and immune evasion ability of biological constituents. The biohybrid delivery vectors offer an excellent opportunity to harness the synergistic properties of the best entities of the two worlds for improved therapeutic outputs. The major spotlights of this review are different biological components, synthetic counterparts of biohybrid nanocarriers, recent advances in hybridization techniques, and the design of biohybrid delivery systems for cancer therapy. Moreover, this review provides an overview of biohybrid systems with therapeutic and diagnostic applications. In a nutshell, this article summarizes the advantages and limitations of various biohybrid nano-platforms, their clinical potential and future directions for successful translation in cancer management.
Collapse
Affiliation(s)
- Mayur Aalhate
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, (NIPER), Hyderabad 500037, India
| | - Srushti Mahajan
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, (NIPER), Hyderabad 500037, India
| | - Anish Dhuri
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, (NIPER), Hyderabad 500037, India
| | - Pankaj Kumar Singh
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, (NIPER), Hyderabad 500037, India.
| |
Collapse
|
4
|
Cardellini J, Normak K, Gerlt M, Makasewicz K, Seiffert C, Capasso Palmiero U, Ye S, González Gómez MA, Piñero Y, Rivas J, Bongiovanni A, Bergese P, Arosio P. Microfluidics-Driven Manufacturing and Multiscale Analytical Characterization of Nanoparticle-Vesicle Hybrids. Adv Healthc Mater 2024:e2403264. [PMID: 39722148 DOI: 10.1002/adhm.202403264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 12/04/2024] [Indexed: 12/28/2024]
Abstract
Coating synthetic nanoparticles (NPs) with lipid membranes is a promising approach to enhance the performance of nanomaterials in various biological applications, including therapeutic delivery to target organs. Current methods for achieving this coating often rely on bulk approaches which can result in low efficiency and poor reproducibility. Continuous processes coupled with quality control represent an attractive strategy to manufacture products with consistent attributes and high yields. Here, this concept is implemented by developing an acoustic microfluidic device together with an analytical platform to prepare nanoparticle-vesicle hybrids and quantitatively characterize the nanoparticle coverage using fluorescence-based techniques at different levels of resolution. With this approach polymethyl methacrylate (PMMA) nanoparticles are successfully coated with liposomes and extracellular vesicles (EVs), achieving a high encapsulation efficiency of 70%. Moreover, the approach enables the identification of design rules to control the efficiency of encapsulation by tuning various operational parameters and material properties, including buffer composition, nanoparticle/vesicle ratio, and vesicle rigidity.
Collapse
Affiliation(s)
- Jacopo Cardellini
- ETH Zürich, Department of Chemistry and Applied Biosciences, Institute for Chemical and Bioengineering, 8093, Zürich, Switzerland
- Department of Chemistry "Ugo Schiff," University of Florence, 50019 Florence, Italy
| | - Karl Normak
- ETH Zürich, Department of Chemistry and Applied Biosciences, Institute for Chemical and Bioengineering, 8093, Zürich, Switzerland
| | - Michael Gerlt
- ETH Zürich, Department of Chemistry and Applied Biosciences, Institute for Chemical and Bioengineering, 8093, Zürich, Switzerland
| | - Katarzyna Makasewicz
- ETH Zürich, Department of Chemistry and Applied Biosciences, Institute for Chemical and Bioengineering, 8093, Zürich, Switzerland
| | - Charlotte Seiffert
- ETH Zürich, Department of Chemistry and Applied Biosciences, Institute for Chemical and Bioengineering, 8093, Zürich, Switzerland
| | - Umberto Capasso Palmiero
- ETH Zürich, Department of Chemistry and Applied Biosciences, Institute for Chemical and Bioengineering, 8093, Zürich, Switzerland
| | - Suiying Ye
- ETH Zürich, Department of Chemistry and Applied Biosciences, Institute for Chemical and Bioengineering, 8093, Zürich, Switzerland
| | - Manuel A González Gómez
- Nanotechnology and Magnetism Lab - NANOMAG, Materials Institute - iMATUS, Health Research Institute - IDIS, Department of Applied Physics, Universidade de Santiago de Compostela, 15782, Santiago, Spain
| | - Yolanda Piñero
- Nanostructured Materials Group, International Iberian Nanotechnology Laboratory (INL), Avenida Mestre Jose Veiga, Braga, 4715-330, Portugal
| | - José Rivas
- Nanotechnology and Magnetism Lab - NANOMAG, Materials Institute - iMATUS, Health Research Institute - IDIS, Department of Applied Physics, Universidade de Santiago de Compostela, 15782, Santiago, Spain
| | - Antonella Bongiovanni
- Cell-Tech HUB at Institute for Research and Biomedical Innovation, National Research Council of Italy (CNR), 90146 Palermo, Italy
| | - Paolo Bergese
- Department of Molecular and Translational Medicine, Università degli Studi di Brescia, Viale Europa 11, 25123, Brescia, Italy
- Center for Colloid and Surface Science (CSGI), Via della Lastruccia 3, 50019, Sesto Fiorentino, Firenze, Italy
| | - Paolo Arosio
- ETH Zürich, Department of Chemistry and Applied Biosciences, Institute for Chemical and Bioengineering, 8093, Zürich, Switzerland
| |
Collapse
|
5
|
Yu S, Rejinold NS, Choi G, Choy JH. Revolutionizing healthcare: inorganic medicinal nanoarchitectonics for advanced theranostics. NANOSCALE HORIZONS 2024. [PMID: 39648727 DOI: 10.1039/d4nh00497c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/10/2024]
Abstract
Over the last two decades, advancements in nanomaterials and nanoscience have paved the path for the emergence of nano-medical convergence science, significantly impacting healthcare. In our review, we highlight how these advancements are applied in various biomedical technologies such as drug delivery systems, bio-imaging for diagnostic and therapeutic purposes. Recently, novel inorganic nanohybrid drugs have been developed, combining multifunctional inorganic nanomaterials with therapeutic agents (known as inorganic medicinal nanoarchitectonics). These innovative drugs are actively utilized in cutting-edge medical treatments, including targeted anti-cancer therapy, photo and radiation therapy, and immunotherapy. This review provides a detailed overview of the current development status of inorganic medicinal nanoarchitectonics and explores potential future directions in their advancements.
Collapse
Affiliation(s)
- Seungjin Yu
- Intelligent Nanohybrid Materials Laboratory (INML), Department of Chemistry, College of Science and Technology, Dankook University, Cheonan 31116, Republic of Korea.
- Department of Nanobiomedical Science, Dankook University, Cheonan 31116, Republic of Korea
| | - N Sanoj Rejinold
- Intelligent Nanohybrid Materials Laboratory (INML), Department of Chemistry, College of Science and Technology, Dankook University, Cheonan 31116, Republic of Korea.
| | - Goeun Choi
- Intelligent Nanohybrid Materials Laboratory (INML), Department of Chemistry, College of Science and Technology, Dankook University, Cheonan 31116, Republic of Korea.
- Department of Nanobiomedical Science, Dankook University, Cheonan 31116, Republic of Korea
| | - Jin-Ho Choy
- Intelligent Nanohybrid Materials Laboratory (INML), Department of Chemistry, College of Science and Technology, Dankook University, Cheonan 31116, Republic of Korea.
- Division of Natural Sciences, The National Academy of Sciences, Seoul 06579, Republic of Korea
- Tokyo Tech Tokyo Tech World Research Hub Initiative (WRHI), Institute of Innovative Research, Institute of Science Tokyo, Yokohama 226853, Japan
| |
Collapse
|
6
|
Zhu Y, Zhao J, Ding H, Qiu M, Xue L, Ge D, Wen G, Ren H, Li P, Wang J. Applications of plant-derived extracellular vesicles in medicine. MedComm (Beijing) 2024; 5:e741. [PMID: 39309692 PMCID: PMC11413507 DOI: 10.1002/mco2.741] [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: 05/28/2024] [Revised: 08/28/2024] [Accepted: 08/28/2024] [Indexed: 09/25/2024] Open
Abstract
Plant-derived extracellular vesicles (EVs) are promising therapeutic agents owing to their natural abundance, accessibility, and unique biological properties. This review provides a comprehensive exploration of the therapeutic potential of plant-derived EVs and emphasizes their anti-inflammatory, antimicrobial, and tumor-inhibitory effects. Here, we discussed the advancements in isolation and purification techniques, such as ultracentrifugation and size-exclusion chromatography, which are critical for maintaining the functional integrity of these nanovesicles. Next, we investigated the diverse administration routes of EVs and carefully weighed their respective advantages and challenges related to bioavailability and patient compliance. Moreover, we elucidated the multifaceted mechanisms of action of plant-derived EVs, including their roles in anti-inflammation, antioxidation, antitumor activity, and modulation of gut microbiota. We also discussed the impact of EVs on specific diseases such as cancer and inflammatory bowel disease, highlighting the importance of addressing current challenges related to production scalability, regulatory compliance, and immunogenicity. Finally, we proposed future research directions for optimizing EV extraction and developing targeted delivery systems. Through these efforts, we envision the seamless integration of plant-derived EVs into mainstream medicine, offering safe and potent therapeutic alternatives across various medical disciplines.
Collapse
Affiliation(s)
- Yawen Zhu
- Division of Hepatobiliary and Transplantation SurgeryDepartment of General SurgeryNanjing Drum Tower HospitalClinical College of Nanjing University of Chinese MedicineNanjingChina
| | - Junqi Zhao
- Division of Hepatobiliary and Transplantation SurgeryDepartment of General SurgeryNanjing Drum Tower HospitalClinical College of Nanjing University of Chinese MedicineNanjingChina
| | - Haoran Ding
- Division of Hepatobiliary and Transplantation SurgeryDepartment of General SurgeryNanjing Drum Tower HospitalClinical College of Nanjing University of Chinese MedicineNanjingChina
| | - Mengdi Qiu
- Division of Hepatobiliary and Transplantation SurgeryDepartment of General SurgeryNanjing Drum Tower HospitalClinical College of Nanjing University of Chinese MedicineNanjingChina
| | - Lingling Xue
- Division of Hepatobiliary and Transplantation SurgeryDepartment of General SurgeryNanjing Drum Tower HospitalClinical College of Nanjing University of Chinese MedicineNanjingChina
| | - Dongxue Ge
- Division of Hepatobiliary and Transplantation SurgeryDepartment of General SurgeryNanjing Drum Tower HospitalClinical College of Nanjing University of Chinese MedicineNanjingChina
| | - Gaolin Wen
- Division of Hepatobiliary and Transplantation SurgeryDepartment of General SurgeryNanjing Drum Tower HospitalClinical College of Nanjing University of Chinese MedicineNanjingChina
| | - Haozhen Ren
- Division of Hepatobiliary and Transplantation SurgeryDepartment of General SurgeryNanjing Drum Tower HospitalClinical College of Nanjing University of Chinese MedicineNanjingChina
| | - Peng Li
- Department of CardiologyThe First Affiliated Hospital of Nanjing Medical UniversityNanjingJiangsuChina
| | - Jinglin Wang
- Division of Hepatobiliary and Transplantation SurgeryDepartment of General SurgeryNanjing Drum Tower HospitalClinical College of Nanjing University of Chinese MedicineNanjingChina
| |
Collapse
|
7
|
Tripathi AD, Labh Y, Katiyar S, Chaturvedi VK, Sharma P, Mishra A. Advancements in Nano-Mediated Biosensors: Targeting Cancer Exosome Detection. J CLUST SCI 2024; 35:2195-2212. [DOI: 10.1007/s10876-024-02676-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 07/24/2024] [Indexed: 01/05/2025]
|
8
|
Chen YW, Lin YH, Ho CC, Chen CY, Yu MH, Lee AKX, Chiu SC, Cho DY, Shie MY. High-yield extracellular vesicle production from HEK293T cells encapsulated in 3D auxetic scaffolds with cyclic mechanical stimulation for effective drug carrier systems. Biofabrication 2024; 16:045035. [PMID: 39173665 DOI: 10.1088/1758-5090/ad728b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Accepted: 08/22/2024] [Indexed: 08/24/2024]
Abstract
Extracellular vesicles (EVs) show promise in drug loading and delivery for medical applications. However, the lack of scalable manufacturing processes hinders the generation of clinically suitable quantities, thereby impeding the translation of EV-based therapies. Current EV production relies heavily on non-physiological two-dimensional (2D) cell culture or bioreactors, requiring significant resources. Additionally, EV-derived ribonucleic acid cargo in three-dimensional (3D) and 2D culture environments remains largely unknown. In this study, we optimized the biofabrication of 3D auxetic scaffolds encapsulated with human embryonic kidney 293 T (HEK293 T) cells, focusing on enhancing the mechanical properties of the scaffolds to significantly boost EV production through tensile stimulation in bioreactors. The proposed platform increased EV yields approximately 115-fold compared to conventional 2D culture, possessing properties that inhibit tumor progression. Further mechanistic examinations revealed that this effect was mediated by the mechanosensitivity of YAP/TAZ. EVs derived from tensile-stimulated HEK293 T cells on 3D auxetic scaffolds demonstrated superior capability for loading doxorubicin compared to their 2D counterparts for cancer therapy. Our results underscore the potential of this strategy for scaling up EV production and optimizing functional performance for clinical translation.
Collapse
Affiliation(s)
- Yi-Wen Chen
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 406040, Taiwan
- Research & Development Center for x-Dimensional Extracellular Vesicles, China Medical University Hospital, Taichung 404332, Taiwan
| | - Yen-Hong Lin
- Research & Development Center for x-Dimensional Extracellular Vesicles, China Medical University Hospital, Taichung 404332, Taiwan
- Department of Biomedical Engineering, China Medical University, Taichung 406040, Taiwan
| | - Chia-Che Ho
- Department of Bioinformatics and Medical Engineering, Asia University, Taichung 41354, Taiwan
- High Performance Materials Institute for x-Dimensional Printing, Asia University, Taichung 41354, Taiwan
| | - Cheng-Yu Chen
- Research & Development Center for x-Dimensional Extracellular Vesicles, China Medical University Hospital, Taichung 404332, Taiwan
| | - Min-Hua Yu
- Institute of Translational Medicine and New Drug Development, China Medical University, Taichung 406040, Taiwan
| | - Alvin Kai-Xing Lee
- Department of Orthopedics, China Medical University Hospital, Taichung 404332, Taiwan
| | - Shao-Chih Chiu
- Translational Cell Therapy Center, China Medical University Hospital, Taichung 404332, Taiwan
| | - Der-Yang Cho
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 406040, Taiwan
- Research & Development Center for x-Dimensional Extracellular Vesicles, China Medical University Hospital, Taichung 404332, Taiwan
- Translational Cell Therapy Center, China Medical University Hospital, Taichung 404332, Taiwan
- Department of Neurosurgery, China Medical University Hospital, Taichung 404332, Taiwan
| | - Ming-You Shie
- Research & Development Center for x-Dimensional Extracellular Vesicles, China Medical University Hospital, Taichung 404332, Taiwan
- Department of Biomedical Engineering, China Medical University, Taichung 406040, Taiwan
- Department of Bioinformatics and Medical Engineering, Asia University, Taichung 41354, Taiwan
| |
Collapse
|
9
|
Zeynalzadeh E, Khodadadi E, Khodadadi E, Ahmadian Z, Kazeminava F, Rasoulzadehzali M, Samadi Kafil H. Navigating the neurological frontier: Macromolecular marvels in overcoming blood-brain barrier challenges for advanced drug delivery. Heliyon 2024; 10:e35562. [PMID: 39170552 PMCID: PMC11336773 DOI: 10.1016/j.heliyon.2024.e35562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 07/29/2024] [Accepted: 07/31/2024] [Indexed: 08/23/2024] Open
Abstract
The blood-brain interface poses formidable obstacles in addressing neurological conditions such as Alzheimer's, Multiple Sclerosis, brain cancers, and cerebrovascular accidents. Serving as a safeguard against potential threats in the blood, this barrier hinders direct drug delivery to affected cells, necessitating specialized transport mechanisms. Within the realm of nanotechnology, the creation of nanoscale carriers, including macromolecules such as polymers, lipids, and metallic nanoparticles, is gaining prominence. These carriers, tailored in diverse forms and sizes and enriched with specific functional groups for enhanced penetration and targeting, are capturing growing interest. This revised abstract explores the macromolecular dimension in understanding how nanoparticles interact with the blood-brain barrier. It re-evaluates the structure and function of the blood-brain barrier, highlighting macromolecular nanocarriers utilized in drug delivery to the brain. The discussion delves into the intricate pathways through which drugs navigate the blood-brain barrier, emphasizing the distinctive attributes of macromolecular nanocarriers. Additionally, it explores recent innovations in nanotechnology and unconventional approaches to drug delivery. Ultimately, the paper addresses the intricacies and considerations in developing macromolecular-based nanomedicines for the brain, aiming to advance the creation and evolution of nanomedicines for neurological ailments.
Collapse
Affiliation(s)
- Elham Zeynalzadeh
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ehsan Khodadadi
- Drugs Applied Research Center, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ehsaneh Khodadadi
- Drugs Applied Research Center, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Zainab Ahmadian
- Department of Pharmaceutics, School of Pharmacy, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Fahimeh Kazeminava
- Drugs Applied Research Center, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Monireh Rasoulzadehzali
- Drugs Applied Research Center, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hossein Samadi Kafil
- Drugs Applied Research Center, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
10
|
Wang J, Yin B, Lian J, Wang X. Extracellular Vesicles as Drug Delivery System for Cancer Therapy. Pharmaceutics 2024; 16:1029. [PMID: 39204374 PMCID: PMC11359799 DOI: 10.3390/pharmaceutics16081029] [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: 06/17/2024] [Revised: 07/18/2024] [Accepted: 07/29/2024] [Indexed: 09/04/2024] Open
Abstract
In recent decades, the pursuit of drug delivery systems has led to the development of numerous synthetic options aimed at enhancing drug efficacy while minimizing side effects. However, the practical application of these systems is often hindered by challenges such as inefficiency, cytotoxicity, and immunogenicity. Extracellular vesicles, natural carriers for drugs, emerge as promising alternatives with distinct advantages over synthetic carriers. Notably, EVs exhibit biocompatibility, low immunogenicity, and inherent tissue-targeting capabilities, thus opening new avenues for drug delivery strategies. This review provides an overview of EVs, including their biogenesis and absorption mechanisms. Additionally, we explore the current research efforts focusing on harnessing their potential as drug carriers, encompassing aspects such as purification techniques, drug loading, and bioengineering for targeted delivery. Finally, we discuss the existing challenges and future prospects of EVs as therapeutic agents in clinical settings. This comprehensive analysis aims to shed light on the potential of EVs as versatile and effective tools for drug delivery, particularly in the realm of cancer therapy.
Collapse
Affiliation(s)
- Jin Wang
- School of Life Sciences, Liaoning University, Shenyang 110036, China; (J.W.); (J.L.)
| | - Bohang Yin
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University, Shenyang 110001, China;
| | - Jiabing Lian
- School of Life Sciences, Liaoning University, Shenyang 110036, China; (J.W.); (J.L.)
| | - Xia Wang
- Institute of Health Sciences, China Medical University, 77 Puhe Road, Shenyang 110122, China
| |
Collapse
|
11
|
You QY, Hu MD, Qian H. Advanced Nanoarchitectonics of Drug Delivery Systems with Pyroptosis Inhibition for Noncancerous Disease Treatment. ADVANCED FUNCTIONAL MATERIALS 2024; 34. [DOI: 10.1002/adfm.202315199] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Indexed: 01/06/2025]
Abstract
AbstractProgrammed cell death (PCD) is a controlled and organized form of death regulated by genes, allowing cells to adapt to their environment. Pyroptosis, a recently discovered type of programmed cell death, differs from apoptosis and necrosis. It is characterized by the activation of caspase and the cleavage of gasdermin. Many studies have focused on understanding the mechanisms and roles of pyroptosis, particularly in cancer research. While inducing pyroptosis in tumor cells for cancer treatment is a major research focus, it is equally important to explore methods of reducing pyroptosis in noncancerous diseases. Recent advancements in drug delivery systems, specifically nanoarchitectonics, offer site‐specific targeting, prolonged drug circulation, enhanced efficacy, improved solubility, and better absorption. Although several reviews have described how nanoarchitectonics can trigger pyroptosis in tumor cells, little attention is given to their potential to inhibit pyroptosis in noncancerous diseases. Therefore, it is crucial to bridge this gap and explore the future directions for utilizing nanoarchitectonics as a powerful tool against noncancerous diseases. This review aims to delve into the recent progress made in nanoarchitectonics‐based advanced drug delivery systems for the treatment of noncancerous diseases by reducing pyroptosis, while also highlighting potential future perspectives in this emerging field.
Collapse
Affiliation(s)
- Qian Yi You
- Department of Geriatrics and Special Services Medicine Xinqiao Hospital of Army Medical University (Third Military Medical University) 183 Xinqiao Street Chongqing 400037 P. R. China
- Institute of Respiratory Diseases Xinqiao Hospital of Army Medical University (Third Military Medical University) 183 Xinqiao Street Chongqing 400037 P. R. China
| | - Ming Dong Hu
- Department of Geriatrics and Special Services Medicine Xinqiao Hospital of Army Medical University (Third Military Medical University) 183 Xinqiao Street Chongqing 400037 P. R. China
| | - Hang Qian
- Institute of Respiratory Diseases Xinqiao Hospital of Army Medical University (Third Military Medical University) 183 Xinqiao Street Chongqing 400037 P. R. China
| |
Collapse
|
12
|
Ariga K, Song J, Kawakami K. Molecular machines working at interfaces: physics, chemistry, evolution and nanoarchitectonics. Phys Chem Chem Phys 2024; 26:13532-13560. [PMID: 38654597 DOI: 10.1039/d4cp00724g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
As a post-nanotechnology concept, nanoarchitectonics combines nanotechnology with advanced materials science. Molecular machines made by assembling molecular units and their organizational bodies are also products of nanoarchitectonics. They can be regarded as the smallest functional materials. Originally, studies on molecular machines analyzed the average properties of objects dispersed in solution by spectroscopic methods. Researchers' playgrounds partially shifted to solid interfaces, because high-resolution observation of molecular machines is usually done on solid interfaces under high vacuum and cryogenic conditions. Additionally, to ensure the practical applicability of molecular machines, operation under ambient conditions is necessary. The latter conditions are met in dynamic interfacial environments such as the surface of water at room temperature. According to these backgrounds, this review summarizes the trends of molecular machines that continue to evolve under the concept of nanoarchitectonics in interfacial environments. Some recent examples of molecular machines in solution are briefly introduced first, which is followed by an overview of studies of molecular machines and similar supramolecular structures in various interfacial environments. The interfacial environments are classified into (i) solid interfaces, (ii) liquid interfaces, and (iii) various material and biological interfaces. Molecular machines are expanding their activities from the static environment of a solid interface to the more dynamic environment of a liquid interface. Molecular machines change their field of activity while maintaining their basic functions and induce the accumulation of individual molecular machines into macroscopic physical properties molecular machines through macroscopic mechanical motions can be employed to control molecular machines. Moreover, research on molecular machines is not limited to solid and liquid interfaces; interfaces with living organisms are also crucial.
Collapse
Affiliation(s)
- Katsuhiko Ariga
- Research Center for Materials Nanoarchitectonics, National Institute for Materials Science (NIMS), 1-1 Namiki, Tsukuba 305-0044, Japan.
- Graduate School of Frontier Sciences, The University of Tokyo, 5-1-5 Kashiwa-no-ha, Kashiwa 277-8561, Japan
| | - Jingwen Song
- Research Center for Functional Materials, National Institute for Materials Science (NIMS), 1-1 Namiki, Tsukuba 305-0044, Ibaraki, Japan
| | - Kohsaku Kawakami
- Research Center for Functional Materials, National Institute for Materials Science (NIMS), 1-1 Namiki, Tsukuba 305-0044, Ibaraki, Japan
- Graduate School of Pure and Applied Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba 305-8577, Ibaraki, Japan
| |
Collapse
|
13
|
Tang J, Wang X, Lin X, Wu C. Mesenchymal stem cell-derived extracellular vesicles: a regulator and carrier for targeting bone-related diseases. Cell Death Discov 2024; 10:212. [PMID: 38697996 PMCID: PMC11066013 DOI: 10.1038/s41420-024-01973-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 04/16/2024] [Accepted: 04/17/2024] [Indexed: 05/05/2024] Open
Abstract
The escalating threat of bone-related diseases poses a significant challenge to human health. Mesenchymal stem cell (MSC)-derived extracellular vesicles (MSC-EVs), as inherent cell-secreted natural products, have emerged as promising treatments for bone-related diseases. Leveraging outstanding features such as high biocompatibility, low immunogenicity, superior biological barrier penetration, and extended circulating half-life, MSC-EVs serve as potent carriers for microRNAs (miRNAs), long no-code RNAs (lncRNAs), and other biomolecules. These cargo molecules play pivotal roles in orchestrating bone metabolism and vascularity through diverse mechanisms, thereby contributing to the amelioration of bone diseases. Additionally, engineering modifications enhance the bone-targeting ability of MSC-EVs, mitigating systemic side effects and bolstering their clinical translational potential. This review comprehensively explores the mechanisms through which MSC-EVs regulate bone-related disease progression. It delves into the therapeutic potential of MSC-EVs as adept drug carriers, augmented by engineered modification strategies tailored for osteoarthritis (OA), rheumatoid arthritis (RA), osteoporosis, and osteosarcoma. In conclusion, the exceptional promise exhibited by MSC-EVs positions them as an excellent solution with considerable translational applications in clinical orthopedics.
Collapse
Affiliation(s)
- Jiandong Tang
- Orthopaedics Center, Zigong Fourth People's Hospital, Tan mu lin Street 19#, Zigong, 643099, Sichuan Province, China
| | - Xiangyu Wang
- Orthopaedics Center, Zigong Fourth People's Hospital, Tan mu lin Street 19#, Zigong, 643099, Sichuan Province, China
| | - Xu Lin
- Orthopaedics Center, Zigong Fourth People's Hospital, Tan mu lin Street 19#, Zigong, 643099, Sichuan Province, China
| | - Chao Wu
- Orthopaedics Center, Zigong Fourth People's Hospital, Tan mu lin Street 19#, Zigong, 643099, Sichuan Province, China.
| |
Collapse
|
14
|
Cheng LF, You CQ, Peng C, Ren JJ, Guo K, Liu TL. Mesenchymal stem cell-derived exosomes as a new drug carrier for the treatment of spinal cord injury: A review. Chin J Traumatol 2024; 27:134-146. [PMID: 38570272 PMCID: PMC11138942 DOI: 10.1016/j.cjtee.2024.03.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 02/20/2024] [Accepted: 03/15/2024] [Indexed: 04/05/2024] Open
Abstract
Spinal cord injury (SCI) is a devastating traumatic disease seriously impairing the quality of life in patients. Expectations to allow the hopeless central nervous system to repair itself after injury are unfeasible. Developing new approaches to regenerate the central nervous system is still the priority. Exosomes derived from mesenchymal stem cells (MSC-Exo) have been proven to robustly quench the inflammatory response or oxidative stress and curb neuronal apoptosis and autophagy following SCI, which are the key processes to rescue damaged spinal cord neurons and restore their functions. Nonetheless, MSC-Exo in SCI received scant attention. In this review, we reviewed our previous work and other studies to summarize the roles of MSC-Exo in SCI and its underlying mechanisms. Furthermore, we also focus on the application of exosomes as drug carrier in SCI. In particular, it combs the advantages of exosomes as a drug carrier for SCI, imaging advantages, drug types, loading methods, etc., which provides the latest progress for exosomes in the treatment of SCI, especially drug carrier.
Collapse
Affiliation(s)
- Lin-Fei Cheng
- Medical College, Anhui University of Science and Technology, Huainan, 232000, Anhui province, China
| | - Chao-Qun You
- Department of Orthopaedic Oncology, Changzheng Hospital, Navy Medical University, Shanghai, 200003, China
| | - Cheng Peng
- Department of Orthopaedic Oncology, Changzheng Hospital, Navy Medical University, Shanghai, 200003, China
| | - Jia-Ji Ren
- Department of Orthopaedic Oncology, Changzheng Hospital, Navy Medical University, Shanghai, 200003, China
| | - Kai Guo
- Department of Orthopaedics, The Central Hospital of Shanghai Putuo District, Shanghai, 200333, China
| | - Tie-Long Liu
- Medical College, Anhui University of Science and Technology, Huainan, 232000, Anhui province, China.
| |
Collapse
|
15
|
Chu X, Hou M, Li Y, Zhang Q, Wang S, Ma J. Extracellular vesicles in endometriosis: role and potential. Front Endocrinol (Lausanne) 2024; 15:1365327. [PMID: 38737555 PMCID: PMC11082332 DOI: 10.3389/fendo.2024.1365327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 04/11/2024] [Indexed: 05/14/2024] Open
Abstract
Endometriosis is a chronic inflammatory gynecological disease, which profoundly jeopardizes women's quality of life and places a significant medical burden on society. The pathogenesis of endometriosis remains unclear, posing major clinical challenges in diagnosis and treatment. There is an urgent demand for the development of innovative non-invasive diagnostic techniques and the identification of therapeutic targets. Extracellular vesicles, recognized for transporting a diverse array of signaling molecules, have garnered extensive attention as a novel mode of intercellular communication. A burgeoning body of research indicates that extracellular vesicles play a pivotal role in the pathogenesis of endometriosis, which may provide possibility and prospect for both diagnosis and treatment. In light of this context, this article focuses on the involvement of extracellular vesicles in the pathogenesis of endometriosis, which deliver information among endometrial stromal cells, macrophages, mesenchymal stem cells, and other cells, and explores their potential applications in the diagnosis and treatment, conducing to the emergence of new strategies for clinical diagnosis and treatment.
Collapse
Affiliation(s)
| | | | | | | | | | - Jing Ma
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| |
Collapse
|
16
|
Yang LY, Li CQ, Zhang YL, Ma MW, Cheng W, Zhang GJ. Emerging Drug Delivery Vectors: Engineering of Plant-Derived Nanovesicles and Their Applications in Biomedicine. Int J Nanomedicine 2024; 19:2591-2610. [PMID: 38505167 PMCID: PMC10949304 DOI: 10.2147/ijn.s454794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 02/28/2024] [Indexed: 03/21/2024] Open
Abstract
Extracellular vesicles can transmit intercellular information and transport biomolecules to recipient cells during various pathophysiological processes in the organism. Animal cell exosomes have been identified as potential nanodrugs delivery vehicles, yet they have some shortcomings such as high immunogenicity, high cytotoxicity, and complicated preparation procedures. In addition to exosomes, plant-derived extracellular vesicles (PDVs), which carry a variety of active substances, are another promising nano-transport vehicles emerging in recent years due to their stable physicochemical properties, wide source, and low cost. This work briefly introduces the collection and characterization of PDVs, then focuses on the application of PDVs as natural or engineered drug carriers in biomedicine, and finally discusses the development and challenges of PDVs in future applications.
Collapse
Affiliation(s)
- Lu-Yao Yang
- School of Laboratory Medicine, Hubei University of Chinese Medicine, Wuhan, 430065, People’s Republic of China
| | - Chao-Qing Li
- School of Laboratory Medicine, Hubei University of Chinese Medicine, Wuhan, 430065, People’s Republic of China
- Hubei Shizhen Laboratory, Wuhan, 430065, People’s Republic of China
| | - Yu-Lin Zhang
- School of Laboratory Medicine, Hubei University of Chinese Medicine, Wuhan, 430065, People’s Republic of China
- Hubei Shizhen Laboratory, Wuhan, 430065, People’s Republic of China
| | - Meng-Wen Ma
- Britton Chance Center for Biomedical Photonics at Wuhan National Laboratory for Optoelectronics - Hubei Bioinformatics & Molecular Imaging Key Laboratory, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, People’s Republic of China
| | - Wan Cheng
- Britton Chance Center for Biomedical Photonics at Wuhan National Laboratory for Optoelectronics - Hubei Bioinformatics & Molecular Imaging Key Laboratory, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, People’s Republic of China
| | - Guo-Jun Zhang
- School of Laboratory Medicine, Hubei University of Chinese Medicine, Wuhan, 430065, People’s Republic of China
- Hubei Shizhen Laboratory, Wuhan, 430065, People’s Republic of China
| |
Collapse
|
17
|
Zhang RY, Cheng K, Huang ZY, Zhang XS, Li Y, Sun X, Yang XQ, Hu YG, Hou XL, Liu B, Chen W, Fan JX, Zhao YD. M1 macrophage-derived exosome for reprograming M2 macrophages and combining endogenous NO gas therapy with enhanced photodynamic synergistic therapy in colorectal cancer. J Colloid Interface Sci 2024; 654:612-625. [PMID: 37862809 DOI: 10.1016/j.jcis.2023.10.054] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 09/19/2023] [Accepted: 10/12/2023] [Indexed: 10/22/2023]
Abstract
Reprogramming immunosuppressive M2 macrophages into M1 macrophages in tumor site provides a new strategy for the immunotherapy of colorectal cancer. In this study, M1 macrophage-derived exosome nanoprobe (M1UC) with Ce6-loaded upconversion material is designed to enhance the photodynamic performance of Ce6 while reprogramming M2 macrophages at tumor site and producing NO gas for three-mode synergistic therapy. Under the excitation of near-infrared light at 808 nm, the probe can generate 660 nm up-conversion fluorescence, which enables the photosensitizer Ce6 to produce ROS efficiently. In addition, the probe leads the production of NO by nitric oxide synthase on exosomes. Confocal laser and flow cytometry results show that M1UC probe reprograms M2 macrophages into M1 macrophages with an efficiency of 95.12%. The cell experiments show that the apoptosis rate of the three-mode synergistic therapy group is 78.8%, and the therapeutic effect is significantly higher than those of the other single treatment groups. In vivo experiments results show that M1UC probes maximally gather at the tumor site after 12 h of intravenous injection in orthotopic colorectal cancer mice. After 808 nm laser irradiation, the survival rate of mice is 100% and the recurrence rate was 0 within 60 d, and the therapeutic effect is significantly higher than those of other single treatment groups, which is also confirmed by immunohistochemistry. This M1 macrophage-derived exosome nanoplatform which is based on the three modes of immunotherapy, gas therapy and photodynamic therapy, provides a new design idea for the diagnosis and treatment of deep tumors.
Collapse
Affiliation(s)
- Ruo-Yun Zhang
- Britton Chance Center for Biomedical Photonics at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics & Molecular Imaging Key Laboratory, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, Hubei, PR China; School of Bioengineering and Health, Wuhan Textile University, Wuhan 430200, Hubei, PR China; State Key Laboratory of New Textile Materials and Advanced Processing Technologies, Wuhan Textile University, Wuhan 430200, Hubei, PR China
| | - Kai Cheng
- Britton Chance Center for Biomedical Photonics at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics & Molecular Imaging Key Laboratory, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, Hubei, PR China
| | - Zhuo-Yao Huang
- Britton Chance Center for Biomedical Photonics at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics & Molecular Imaging Key Laboratory, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, Hubei, PR China
| | - Xiao-Shuai Zhang
- Britton Chance Center for Biomedical Photonics at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics & Molecular Imaging Key Laboratory, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, Hubei, PR China; Basic Medical Laboratory, General Hospital of Central Theater Command, Wuhan 430081, Hubei, PR China
| | - Yong Li
- Britton Chance Center for Biomedical Photonics at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics & Molecular Imaging Key Laboratory, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, Hubei, PR China
| | - Xing Sun
- Britton Chance Center for Biomedical Photonics at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics & Molecular Imaging Key Laboratory, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, Hubei, PR China
| | - Xiao-Quan Yang
- Britton Chance Center for Biomedical Photonics at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics & Molecular Imaging Key Laboratory, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, Hubei, PR China
| | - Yong-Guo Hu
- Britton Chance Center for Biomedical Photonics at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics & Molecular Imaging Key Laboratory, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, Hubei, PR China
| | - Xiao-Lin Hou
- Britton Chance Center for Biomedical Photonics at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics & Molecular Imaging Key Laboratory, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, Hubei, PR China
| | - Bo Liu
- Britton Chance Center for Biomedical Photonics at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics & Molecular Imaging Key Laboratory, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, Hubei, PR China
| | - Wei Chen
- Britton Chance Center for Biomedical Photonics at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics & Molecular Imaging Key Laboratory, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, Hubei, PR China
| | - Jin-Xuan Fan
- Britton Chance Center for Biomedical Photonics at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics & Molecular Imaging Key Laboratory, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, Hubei, PR China.
| | - Yuan-Di Zhao
- Britton Chance Center for Biomedical Photonics at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics & Molecular Imaging Key Laboratory, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, Hubei, PR China.
| |
Collapse
|
18
|
Zhang Z, Peng Y, Peng X, Xiao D, Shi Y, Tao Y. Effects of radiation therapy on tumor microenvironment: an updated review. Chin Med J (Engl) 2023; 136:2802-2811. [PMID: 37442768 PMCID: PMC10686612 DOI: 10.1097/cm9.0000000000002535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Indexed: 07/15/2023] Open
Abstract
ABSTRACT Cancer is a major threat to human health and causes death worldwide. Research on the role of radiotherapy (RT) in the treatment of cancer is progressing; however, RT not only causes fatal DNA damage to tumor cells, but also affects the interactions between tumor cells and different components of the tumor microenvironment (TME), including immune cells, fibroblasts, macrophages, extracellular matrix, and some soluble products. Some cancer cells can survive radiation and have shown strong resistance to radiation through interaction with the TME. Currently, the complex relationships between the tumor cells and cellular components that play major roles in various TMEs are poorly understood. This review explores the relationship between RT and cell-cell communication in the TME from the perspective of immunity and hypoxia and aims to identify new RT biomarkers and treatment methods in lung cancer to improve the current status of unstable RT effect and provide a theoretical basis for further lung cancer RT sensitization research in the future.
Collapse
Affiliation(s)
- Zewen Zhang
- NHC Key Laboratory of Carcinogenesis, Cancer Research Institute, Central South University, Changsha, Hunan 410078, China
| | - Yuanhao Peng
- Department of Pathology, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, School of Basic Medicine, Central South University, Changsha, Hunan 410078, China
| | - Xin Peng
- Department of Pathology, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, School of Basic Medicine, Central South University, Changsha, Hunan 410078, China
| | - Desheng Xiao
- Department of Pathology, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, School of Basic Medicine, Central South University, Changsha, Hunan 410078, China
| | - Ying Shi
- NHC Key Laboratory of Carcinogenesis, Cancer Research Institute, Central South University, Changsha, Hunan 410078, China
| | - Yongguang Tao
- NHC Key Laboratory of Carcinogenesis, Cancer Research Institute, Central South University, Changsha, Hunan 410078, China
- Department of Pathology, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, School of Basic Medicine, Central South University, Changsha, Hunan 410078, China
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410078, China
- Department of Thoracic Surgery, Hunan Key Laboratory of Early Diagnosis and Precision Therapy in Lung Cancer, Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| |
Collapse
|
19
|
Ghodasara A, Raza A, Wolfram J, Salomon C, Popat A. Clinical Translation of Extracellular Vesicles. Adv Healthc Mater 2023; 12:e2301010. [PMID: 37421185 DOI: 10.1002/adhm.202301010] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 06/03/2023] [Indexed: 07/10/2023]
Abstract
Extracellular vesicles (EVs) occur in a variety of bodily fluids and have gained recent attraction as natural materials due to their bioactive surfaces, internal cargo, and role in intercellular communication. EVs contain various biomolecules, including surface and cytoplasmic proteins; and nucleic acids that are often representative of the originating cells. EVs can transfer content to other cells, a process that is thought to be important for several biological processes, including immune responses, oncogenesis, and angiogenesis. An increased understanding of the underlying mechanisms of EV biogenesis, composition, and function has led to an exponential increase in preclinical and clinical assessment of EVs for biomedical applications, such as diagnostics and drug delivery. Bacterium-derived EV vaccines have been in clinical use for decades and a few EV-based diagnostic assays regulated under Clinical Laboratory Improvement Amendments have been approved for use in single laboratories. Though, EV-based products are yet to receive widespread clinical approval from national regulatory agencies such as the United States Food and Drug Administration (USFDA) and European Medicine Agency (EMA), many are in late-stage clinical trials. This perspective sheds light on the unique characteristics of EVs, highlighting current clinical trends, emerging applications, challenges and future perspectives of EVs in clinical use.
Collapse
Affiliation(s)
- Aayushi Ghodasara
- School of Pharmacy, The University of Queensland, Brisbane, QLD, 4102, Australia
- Translational Extracellular Vesicles in Obstetrics and Gynae-Oncology Group, The University of Queensland Centre for Clinical Research, Royal Brisbane and Women's Hospital, Faculty of Medicine, The University of Queensland, Brisbane, QLD, 4029, Australia
| | - Aun Raza
- School of Pharmacy, The University of Queensland, Brisbane, QLD, 4102, Australia
| | - Joy Wolfram
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, 4072, Australia
- The School of Chemical Engineering, The University of Queensland, Brisbane, QLD, 4072, Australia
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX, 77030, USA
| | - Carlos Salomon
- Translational Extracellular Vesicles in Obstetrics and Gynae-Oncology Group, The University of Queensland Centre for Clinical Research, Royal Brisbane and Women's Hospital, Faculty of Medicine, The University of Queensland, Brisbane, QLD, 4029, Australia
- Department of Research, Postgraduate and Further Education (DIPEC), Falcuty of Health Sciences, University of Alba, Santiago, 8320000, Chile
| | - Amirali Popat
- School of Pharmacy, The University of Queensland, Brisbane, QLD, 4102, Australia
| |
Collapse
|
20
|
Du S, Guan Y, Xie A, Yan Z, Gao S, Li W, Rao L, Chen X, Chen T. Extracellular vesicles: a rising star for therapeutics and drug delivery. J Nanobiotechnology 2023; 21:231. [PMID: 37475025 PMCID: PMC10360328 DOI: 10.1186/s12951-023-01973-5] [Citation(s) in RCA: 51] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 06/29/2023] [Indexed: 07/22/2023] Open
Abstract
Extracellular vesicles (EVs) are nano-sized, natural, cell-derived vesicles that contain the same nucleic acids, proteins, and lipids as their source cells. Thus, they can serve as natural carriers for therapeutic agents and drugs, and have many advantages over conventional nanocarriers, including their low immunogenicity, good biocompatibility, natural blood-brain barrier penetration, and capacity for gene delivery. This review first introduces the classification of EVs and then discusses several currently popular methods for isolating and purifying EVs, EVs-mediated drug delivery, and the functionalization of EVs as carriers. Thereby, it provides new avenues for the development of EVs-based therapeutic strategies in different fields of medicine. Finally, it highlights some challenges and future perspectives with regard to the clinical application of EVs.
Collapse
Affiliation(s)
- Shuang Du
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, 12 Jichang Road, Guangzhou, 510405, China
| | - Yucheng Guan
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, 12 Jichang Road, Guangzhou, 510405, China
| | - Aihua Xie
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, 12 Jichang Road, Guangzhou, 510405, China
| | - Zhao Yan
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, 12 Jichang Road, Guangzhou, 510405, China
| | - Sijia Gao
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Room 6007, N22, Taipa, 999078, Macau SAR, China
| | - Weirong Li
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, 12 Jichang Road, Guangzhou, 510405, China
| | - Lang Rao
- Institute of Biomedical Health Technology and Engineering, Shenzhen Bay Laboratory, Shenzhen, 518132, China.
| | - Xiaojia Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Room 6007, N22, Taipa, 999078, Macau SAR, China.
| | - Tongkai Chen
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, 12 Jichang Road, Guangzhou, 510405, China.
| |
Collapse
|
21
|
Pei Z, Cen J, Zhang X, Gong C, Sun M, Meng W, Mao G, Wan J, Hu B, He X, Xu Q, Han H, Xiao K. MiR-146a-5p delivered by hucMSC extracellular vesicles modulates the inflammatory response to sulfur mustard-induced acute lung injury. Stem Cell Res Ther 2023; 14:149. [PMID: 37254188 DOI: 10.1186/s13287-023-03375-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 05/11/2023] [Indexed: 06/01/2023] Open
Abstract
BACKGROUND Sulfur mustard (SM) is a highly toxic chemical warfare agent that has caused numerous casualties during wars and conflicts in the past century. Specific antidotes or therapeutic strategies are rare due to the complicated mechanism of toxicity, which still awaits elucidation. Clinical data show that acute lung injury (ALI) is responsible for most mortality and morbidity after SM exposure. Extracellular vesicles are natural materials that participate in intercellular communication by delivering various substances and can be modified. In this study, we aim to show that extracellular vesicles derived from human umbilical cord mesenchymal stromal cells (hucMSC-EVs) could exert therapeutic effects on SM-induced ALI, and to explain the underlying mechanism of effects. METHODS MiR-146a-5p contained in hucMSC-EVs may be involved in the process of hucMSC-EVs modulating the inflammatory response to SM-induced ALI. We utilized miR-146a-5p delivered by extracellular vesicles and further modified hucMSCs with a miR-146a-5p mimic or inhibitor to collect miR-146a-5p-overexpressing extracellular vesicles (miR-146a-5p+-EVs) or miR-146a-5p-underexpressing extracellular vesicles (miR-146a-5p--EVs), respectively. Through in vivo and in vitro experiments, we investigated the mechanism. RESULTS The effect of miR-146a-5p+-EVs on improving the inflammatory reaction tied to SM injury was better than that of hucMSC-EVs. We demonstrated that miR-146a-5p delivered by hucMSC-EVs targeted TRAF6 to negatively regulate inflammation in SM-induced ALI models in vitro and in vivo. CONCLUSION In summary, miR-146a-5p delivered by hucMSC-EVs targeted TRAF6, causing hucMSC-EVs to exert anti-inflammatory effects in SM-induced ALI; thus, hucMSC-EVs treatment may be a promising clinical therapeutic after SM exposure.
Collapse
Affiliation(s)
- Zhipeng Pei
- Department of Protective Medicine Against Chemical Agents, Faculty of Naval Medicine, Naval Medical University, Shanghai, 200433, China
| | - Jinfeng Cen
- Department of Protective Medicine Against Chemical Agents, Faculty of Naval Medicine, Naval Medical University, Shanghai, 200433, China
| | - Xinkang Zhang
- Department of Protective Medicine Against Chemical Agents, Faculty of Naval Medicine, Naval Medical University, Shanghai, 200433, China
| | - Chuchu Gong
- Department of Protective Medicine Against Chemical Agents, Faculty of Naval Medicine, Naval Medical University, Shanghai, 200433, China
| | - Mingxue Sun
- Department of Protective Medicine Against Chemical Agents, Faculty of Naval Medicine, Naval Medical University, Shanghai, 200433, China
| | - Wenqi Meng
- Department of Protective Medicine Against Chemical Agents, Faculty of Naval Medicine, Naval Medical University, Shanghai, 200433, China
| | - Guanchao Mao
- Department of Protective Medicine Against Chemical Agents, Faculty of Naval Medicine, Naval Medical University, Shanghai, 200433, China
| | - Jingjing Wan
- Department of Clinical Pharmacy, School of Pharmacy, Naval Medical University, Shanghai, 200433, China
| | - Bingyue Hu
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Xiaowen He
- Origincell Technology Group Co., Ltd., Shanghai, 201203, China
| | - Qingqiang Xu
- Department of Protective Medicine Against Chemical Agents, Faculty of Naval Medicine, Naval Medical University, Shanghai, 200433, China.
| | - Hua Han
- School of Medicine, Tongji University, Shanghai, 200092, China.
| | - Kai Xiao
- Department of Protective Medicine Against Chemical Agents, Faculty of Naval Medicine, Naval Medical University, Shanghai, 200433, China.
| |
Collapse
|
22
|
Imtiaz F, Islam M, Saeed H, Ahmed A, Asghar M, Saleem B, Farooq MA, Khan DH, Peltonen L. Novel phytoniosomes formulation of Tradescantia pallida leaves attenuates diabetes more effectively than pure extract. J Drug Deliv Sci Technol 2023. [DOI: 10.1016/j.jddst.2023.104399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/04/2023]
|
23
|
Dżaman K, Czerwaty K. Extracellular Vesicle-Based Drug Delivery Systems for Head and Neck Squamous Cell Carcinoma: A Systematic Review. Pharmaceutics 2023; 15:pharmaceutics15051327. [PMID: 37242569 DOI: 10.3390/pharmaceutics15051327] [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: 02/19/2023] [Revised: 04/07/2023] [Accepted: 04/21/2023] [Indexed: 05/28/2023] Open
Abstract
It is estimated that there are over 890,000 new cases of head and neck squamous cell carcinoma (HNSCC) worldwide each year, accounting for approximately 5% of all cancer cases. Current treatment options for HNSCC often cause significant side effects and functional impairments, thus there is a challenge to discover more acceptable treatment technologies. Extracellular vesicles (EVs) can be utilized for HNSCC treatment in several ways, for example, for drug delivery, immune modulation, as biomarkers for diagnostics, gene therapy, or tumor microenvironment modulation. This systematic review summarizes new knowledge regarding these options. Articles published up to 11 December 2022, were identified by searching the electronic databases PubMed/MEDLINE, Scopus, Web of Science, and Cochrane. Only full-text original research papers written in English were considered eligible for analysis. The quality of studies was assessed using the Office of Health Assessment and Translation (OHAT) Risk of Bias Rating Tool for Human and Animal Studies, modified for the needs of this review. Of 436 identified records, 18 were eligible and included. It is important to note that the use of EVs as a treatment for HNSCC is still in the early stages of research, so we summarized information on challenges such as EV isolation, purification, and standardization of EV-based therapies in HNSCC.
Collapse
Affiliation(s)
- Karolina Dżaman
- Department of Otolaryngology, Centre of Postgraduate Medical Education, Marymoncka 99/103, 01-813 Warsaw, Poland
| | - Katarzyna Czerwaty
- Department of Otolaryngology, Centre of Postgraduate Medical Education, Marymoncka 99/103, 01-813 Warsaw, Poland
| |
Collapse
|
24
|
Qiu J, Liu XJ, You BA, Ren N, Liu H. Application of Nanomaterials in Stem Cell-Based Therapeutics for Cardiac Repair and Regeneration. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2206487. [PMID: 36642861 DOI: 10.1002/smll.202206487] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 12/16/2022] [Indexed: 06/17/2023]
Abstract
Cardiovascular disease is a leading cause of disability and death worldwide. Although the survival rate of patients with heart diseases can be improved with contemporary pharmacological treatments and surgical procedures, none of these therapies provide a significant improvement in cardiac repair and regeneration. Stem cell-based therapies are a promising approach for functional recovery of damaged myocardium. However, the available stem cells are difficult to differentiate into cardiomyocytes, which result in the extremely low transplantation efficiency. Nanomaterials are widely used to regulate the myocardial differentiation of stem cells, and play a very important role in cardiac tissue engineering. This study discusses the current status and limitations of stem cells and cell-derived exosomes/micro RNAs based cardiac therapy, describes the cardiac repair mechanism of nanomaterials, summarizes the recent advances in nanomaterials used in cardiac repair and regeneration, and evaluates the advantages and disadvantages of the relevant nanomaterials. Besides discussing the potential clinical applications of nanomaterials in cardiac therapy, the perspectives and challenges of nanomaterials used in stem cell-based cardiac repair and regeneration are also considered. Finally, new research directions in this field are proposed, and future research trends are highlighted.
Collapse
Affiliation(s)
- Jie Qiu
- Medical Research Institute, Jinan Nanjiao Hospital, Jinan, 250002, P. R. China
| | - Xiang-Ju Liu
- Department of Geriatric Medicine, Qilu Hospital of Shandong University, Jinan, 250012, P. R. China
| | - Bei-An You
- Department of Cardiovascular Center, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Jinan, 266035, P. R. China
| | - Na Ren
- Collaborative Innovation Center of Technology and Equipment for Biological Diagnosis and Therapy in Universities of Shandong, Institute for Advanced Interdisciplinary Research (iAIR), School of Chemistry and Chemical Engineering, University of Jinan, Jinan, 250022, P. R. China
| | - Hong Liu
- Collaborative Innovation Center of Technology and Equipment for Biological Diagnosis and Therapy in Universities of Shandong, Institute for Advanced Interdisciplinary Research (iAIR), School of Chemistry and Chemical Engineering, University of Jinan, Jinan, 250022, P. R. China
- State Key Laboratory of Crystal Materials, Shandong University, Jinan, 250100, P. R. China
| |
Collapse
|
25
|
Wu D, Zhang W, Li T, Li F, Feng Q, Cheng X, Guo Y. In situ detection of miRNA-21 in MCF-7 cell-derived extracellular vesicles using the red blood cell membrane vesicle strategy. Chem Commun (Camb) 2023; 59:1987-1990. [PMID: 36723001 DOI: 10.1039/d2cc05954a] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
In this work, we constructed a novel membrane fusion strategy for extracellular vesicles (EVs) and red blood cell membrane vesicles (RVs). A nanoscale space is formed, which can improve the efficiency of the probe reaction with miRNA-21, which allows the in situ fluorescence detection of miRNA-21 in EVs.
Collapse
Affiliation(s)
- Di Wu
- Shandong Provincial Key Laboratory of Molecular Engineering, School of Chemistry and Chemical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan, 250353, China. .,Linyi University, Linyi, 276000, China
| | - Wenyue Zhang
- Shandong Provincial Key Laboratory of Molecular Engineering, School of Chemistry and Chemical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan, 250353, China.
| | - Tao Li
- Shandong Provincial Key Laboratory of Molecular Engineering, School of Chemistry and Chemical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan, 250353, China.
| | - Fen Li
- Shandong Provincial Key Laboratory of Molecular Engineering, School of Chemistry and Chemical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan, 250353, China.
| | - Qingfang Feng
- Shandong Provincial Key Laboratory of Molecular Engineering, School of Chemistry and Chemical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan, 250353, China.
| | - Xiao Cheng
- Shandong Provincial Key Laboratory of Molecular Engineering, School of Chemistry and Chemical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan, 250353, China.
| | - Yingshu Guo
- Shandong Provincial Key Laboratory of Molecular Engineering, School of Chemistry and Chemical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan, 250353, China.
| |
Collapse
|
26
|
Raza A, Rossi GR, Janjua TI, Souza-Fonseca-Guimaraes F, Popat A. Nanobiomaterials to modulate natural killer cell responses for effective cancer immunotherapy. Trends Biotechnol 2023; 41:77-92. [PMID: 35840426 DOI: 10.1016/j.tibtech.2022.06.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 06/08/2022] [Accepted: 06/17/2022] [Indexed: 02/06/2023]
Abstract
Natural killer (NK) cells have emerged as a major target for cancer immunotherapies, particularly as cellular therapy modalities because they have relatively less toxicity than T lymphocytes. However, NK cell-based therapy suffers from many challenges, including problems with its activation, resistance to genetic engineering, and large-scale expansion needed for therapeutic purposes. Recently, nanobiomaterials have emerged as a promising solution to control the challenges associated with NK cells. This focused review summarises the recent advances in the field and highlights current and future perspectives of using nanobiomaterials to maximise anticancer responses of NK cells for safe and effective immunotherapy. Finally, we provide our opinion on the role of smart materials in activating NK cells as a potential cellular therapy of the future.
Collapse
Affiliation(s)
- Aun Raza
- School of Pharmacy, The University of Queensland, Woolloongabba, QLD 4102, Australia
| | - Gustavo Rodrigues Rossi
- University of Queensland Diamantina Institute, The University of Queensland, Woolloongabba, QLD 4102, Australia
| | - Taskeen Iqbal Janjua
- School of Pharmacy, The University of Queensland, Woolloongabba, QLD 4102, Australia
| | | | - Amirali Popat
- School of Pharmacy, The University of Queensland, Woolloongabba, QLD 4102, Australia.
| |
Collapse
|
27
|
Li X, Liu Z, Xu S, Ma X, Zhao Z, Hu H, Deng J, Peng C, Wang Y, Ma S. A drug delivery system constructed by a fusion peptide capturing exosomes targets to titanium implants accurately resulting the enhancement of osseointegration peri-implant. Biomater Res 2022; 26:89. [PMID: 36575503 PMCID: PMC9795642 DOI: 10.1186/s40824-022-00331-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 11/30/2022] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Exosomes derived from bone marrow mesenchymal stem cells (BMSC-exos) have been shown triggering osteogenic differentiation and mineralization of MSCs, but exosomes administered via bolus injections are rapidly sequestered and cleared. Therefore, we considered the implant as a new organ of patient's body and expected to find a method to treat implant with BMSC-exos in vivo directly. METHODS A fusion peptide (PEP), as a drug delivery system (DDS) which contained a titanium-binding peptide (TBP) possessing the ability to selectively bind to the titanium surface and another peptide CP05 being able to capture exosomes expertly, is constructed to modify the titanium surface. RESULTS Both in vitro and in vivo experiments prove PEP retains the ability to bind titanium and exosome simultaneously, and the DDS gain the ability to target exosomes to titanium implants surface following enhancing osseointegration post-implantation. Moreover, the DDS constructed by exosomes of diverse origins shows the similar combination rate and efficiency of therapy. CONCLUSION This drug delivery system demonstrates the concept that EXO-PEP system can offer an accurate and efficient therapy for treating implants with long-term effect.
Collapse
Affiliation(s)
- Xuewen Li
- grid.265021.20000 0000 9792 1228Department of Stomatology, Tianjin Medical University Second Hospital, 23 Pingjiang Road, Tianjin, 300211 China ,grid.265021.20000 0000 9792 1228School and Hospital of Stomotology, Tianjin Medical University, 12 Observatory Road, Heping District, Tianjin, 030070 China
| | - Zihao Liu
- grid.265021.20000 0000 9792 1228School and Hospital of Stomotology, Tianjin Medical University, 12 Observatory Road, Heping District, Tianjin, 030070 China
| | - Shendan Xu
- grid.265021.20000 0000 9792 1228School and Hospital of Stomotology, Tianjin Medical University, 12 Observatory Road, Heping District, Tianjin, 030070 China
| | - Xinying Ma
- grid.265021.20000 0000 9792 1228School and Hospital of Stomotology, Tianjin Medical University, 12 Observatory Road, Heping District, Tianjin, 030070 China
| | - Zhezhe Zhao
- grid.265021.20000 0000 9792 1228School and Hospital of Stomotology, Tianjin Medical University, 12 Observatory Road, Heping District, Tianjin, 030070 China
| | - Han Hu
- grid.265021.20000 0000 9792 1228School and Hospital of Stomotology, Tianjin Medical University, 12 Observatory Road, Heping District, Tianjin, 030070 China
| | - Jiayin Deng
- grid.265021.20000 0000 9792 1228School and Hospital of Stomotology, Tianjin Medical University, 12 Observatory Road, Heping District, Tianjin, 030070 China
| | - Cheng Peng
- grid.265021.20000 0000 9792 1228Department of Stomatology, Tianjin Medical University Second Hospital, 23 Pingjiang Road, Tianjin, 300211 China
| | - Yonglan Wang
- grid.265021.20000 0000 9792 1228School and Hospital of Stomotology, Tianjin Medical University, 12 Observatory Road, Heping District, Tianjin, 030070 China
| | - Shiqing Ma
- grid.265021.20000 0000 9792 1228Department of Stomatology, Tianjin Medical University Second Hospital, 23 Pingjiang Road, Tianjin, 300211 China
| |
Collapse
|
28
|
Wu Y, Chen W, Guo M, Tan Q, Zhou E, Deng J, Li M, Chen J, Yang Z, Jin Y. Metabolomics of Extracellular Vesicles: A Future Promise of Multiple Clinical Applications. Int J Nanomedicine 2022; 17:6113-6129. [PMID: 36514377 PMCID: PMC9741837 DOI: 10.2147/ijn.s390378] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 12/01/2022] [Indexed: 12/12/2022] Open
Abstract
Extracellular vesicles (EVs) can contain DNA, RNA, proteins and metabolic molecules from primary origins; they are coated with a phospholipid bilayer membrane and released by cells into the extracellular matrix. EVs can be obtained from various body liquids, including the blood, saliva, cerebrospinal fluid, and urine. As has been proved, EVs-mediated transfer of biologically active molecules is crucial for various physiological and pathological processes. Extensive investigations have already begun to explore the diagnosis and prognosis potentials for EVs. Furthermore, research has continued to recognize the critical role of nucleic acids and proteins in EVs. However, our understanding of the comprehensive effects of metabolites in these nanoparticles is currently limited and in its infancy. Therefore, we have attempted to summarize the recent research into the metabolomics of EVs in relation to potential clinical applications and discuss the problems and challenges that have occurred, to provide more guidance for the future development in this field.
Collapse
Affiliation(s)
- YaLi Wu
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Respiratory Diseases of National Health Commission, Wuhan Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - WenJuan Chen
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Respiratory Diseases of National Health Commission, Wuhan Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Mengfei Guo
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Respiratory Diseases of National Health Commission, Wuhan Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Qi Tan
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Respiratory Diseases of National Health Commission, Wuhan Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - E Zhou
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Respiratory Diseases of National Health Commission, Wuhan Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Jingjing Deng
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Respiratory Diseases of National Health Commission, Wuhan Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Minglei Li
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Respiratory Diseases of National Health Commission, Wuhan Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Jiangbin Chen
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Respiratory Diseases of National Health Commission, Wuhan Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Zimo Yang
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Respiratory Diseases of National Health Commission, Wuhan Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Yang Jin
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Respiratory Diseases of National Health Commission, Wuhan Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China,Key Laboratory of Biological Targeted Therapy, the Ministry of Education, Wuhan Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China,Clinical Research Center for Major Respiratory Diseases in Hubei Province, Wuhan Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China,Correspondence: Yang Jin, Department of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of Health Ministry, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China, Email
| |
Collapse
|
29
|
Man F, Xing H, Wang H, Wang J, Lu R. Engineered small extracellular vesicles as a versatile platform to efficiently load ferulic acid via an “esterase-responsive active loading” strategy. Front Bioeng Biotechnol 2022; 10:1043130. [DOI: 10.3389/fbioe.2022.1043130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 10/27/2022] [Indexed: 11/11/2022] Open
Abstract
As nano-drug carriers, small extracellular vesicles (sEVs) have shown unique advantages, but their drug loading and encapsulation efficiency are far from being satisfied, especially for the loading of hydrophilic small-molecule drugs. Inspired by the strategies of active loading of liposomal nanomedicines, pre-drug design and immobilization enzyme, here we developed a new platform, named “Esterase-responsive Active Loading” (EAL), for the efficient and stable drug encapsulation of sEVs. Widely used ferulic acid ester derivatives were chosen as prodrugs based on the EAL of engineered sEVs to establish a continuous transmembrane ion gradient for achieving efficient loading of active molecule ferulic acid into sEVs. The EAL showed that the drug loading and encapsulation efficiency were around 6-fold and 5-fold higher than passive loading, respectively. Moreover, characterization by nano-flow cytometry and Malvern particle size analyzer showed that differential ultracentrifugation combined with multiple types of membrane filtration methods can achieve large-scale and high-quality production of sEVs. Finally, extracellular and intracellular assessments further confirmed the superior performance of the EAL-prepared sEVs-loaded ferulic acid preparation in terms of slow release and low toxicity. Taken together, these findings will provide an instructive insight into the development of sEV-based delivery systems.
Collapse
|
30
|
Yu H, Palazzolo JS, Ju Y, Niego B, Pan S, Hagemeyer CE, Caruso F. Polyphenol-Functionalized Cubosomes as Thrombolytic Drug Carriers. Adv Healthc Mater 2022; 11:e2201151. [PMID: 36037807 DOI: 10.1002/adhm.202201151] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Revised: 08/03/2022] [Indexed: 01/28/2023]
Abstract
The safe administration of thrombolytic agents is a challenge for the treatment of acute thrombosis. Lipid-based nanoparticle drug delivery technologies present opportunities to overcome the existing clinical limitations and deliver thrombolytic therapy with enhanced therapeutic outcomes and safety. Herein, lipid cubosomes are examined as nanocarriers for the encapsulation of thrombolytic drugs. The lipid cubosomes are loaded with the thrombolytic drug urokinase-type plasminogen activator (uPA) and coated with a low-fouling peptide that is incorporated within a metal-phenolic network (MPN). The peptide-containing MPN (pep-MPN) coating inhibits the direct contact of uPA with the surrounding environment, as assessed by an in vitro plasminogen activation assay and an ex vivo whole blood clot degradation assay. The pep-MPN-coated cubosomes prepared with 22 wt% peptide demonstrate a cell membrane-dependent thrombolytic activity, which is attributed to their fusogenic lipid behavior. Moreover, compared with the uncoated lipid cubosomes, the uPA-loaded pep-MPN-coated cubosomes demonstrate significantly reduced nonspecific cell association (<10% of the uncoated cubosomes) in the whole blood assay, a prolonged circulating half-life, and reduced splenic uPA accumulation in mice. These studies confirm the preserved bioactivity and cell membrane-dependent release of uPA within pep-MPN-coated lipid cubosomes, highlighting their potential as a delivery vehicle for thrombolytic drugs.
Collapse
Affiliation(s)
- Haitao Yu
- Department of Chemical Engineering, The University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Jason S Palazzolo
- NanoBiotechnology Laboratory, Australian Centre for Blood Diseases, Central Clinical School, Monash University, Melbourne, Victoria, 3004, Australia
| | - Yi Ju
- Department of Chemical Engineering, The University of Melbourne, Parkville, Victoria, 3010, Australia.,School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria, 3083, Australia
| | - Be'eri Niego
- NanoBiotechnology Laboratory, Australian Centre for Blood Diseases, Central Clinical School, Monash University, Melbourne, Victoria, 3004, Australia
| | - Shuaijun Pan
- Department of Chemical Engineering, The University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Christoph E Hagemeyer
- NanoBiotechnology Laboratory, Australian Centre for Blood Diseases, Central Clinical School, Monash University, Melbourne, Victoria, 3004, Australia
| | - Frank Caruso
- Department of Chemical Engineering, The University of Melbourne, Parkville, Victoria, 3010, Australia
| |
Collapse
|
31
|
Lv K, Wang Y, Lou P, Liu S, Zhou P, Yang L, Lu Y, Cheng J, Liu J. Extracellular vesicles as advanced therapeutics for the resolution of organ fibrosis: Current progress and future perspectives. Front Immunol 2022; 13:1042983. [PMCID: PMC9630482 DOI: 10.3389/fimmu.2022.1042983] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 09/30/2022] [Indexed: 11/13/2022] Open
Abstract
Organ fibrosis is a serious health challenge worldwide, and its global incidence and medical burden are increasing dramatically each year. Fibrosis can occur in nearly all major organs and ultimately lead to organ dysfunction. However, current clinical treatments cannot slow or reverse the progression of fibrosis to end-stage organ failure, and thus advanced anti-fibrotic therapeutics are urgently needed. As a type of naturally derived nanovesicle, native extracellular vesicles (EVs) from multiple cell types (e.g., stem cells, immune cells, and tissue cells) have been shown to alleviate organ fibrosis in many preclinical models through multiple effective mechanisms, such as anti-inflammation, pro-angiogenesis, inactivation of myofibroblasts, and fibrinolysis of ECM components. Moreover, the therapeutic potency of native EVs can be further enhanced by multiple engineering strategies, such as genetic modifications, preconditionings, therapeutic reagent-loadings, and combination with functional biomaterials. In this review, we briefly introduce the pathology and current clinical treatments of organ fibrosis, discuss EV biology and production strategies, and particularly focus on important studies using native or engineered EVs as interventions to attenuate tissue fibrosis. This review provides insights into the development and translation of EV-based nanotherapies into clinical applications in the future.
Collapse
Affiliation(s)
- Ke Lv
- National Health Commission (NHC) Key Laboratory of Transplant Engineering and Immunology, Regenerative Medicine Research Center, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Yizhuo Wang
- National Health Commission (NHC) Key Laboratory of Transplant Engineering and Immunology, Regenerative Medicine Research Center, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Peng Lou
- National Health Commission (NHC) Key Laboratory of Transplant Engineering and Immunology, Regenerative Medicine Research Center, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Shuyun Liu
- National Health Commission (NHC) Key Laboratory of Transplant Engineering and Immunology, Regenerative Medicine Research Center, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Pingya Zhou
- National Health Commission (NHC) Key Laboratory of Transplant Engineering and Immunology, Regenerative Medicine Research Center, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Li Yang
- Department of Gastroenterology and Hepatology, Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, West China Hospital, Sichuan University, Chengdu, China
| | - Yanrong Lu
- National Health Commission (NHC) Key Laboratory of Transplant Engineering and Immunology, Regenerative Medicine Research Center, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Jingqiu Cheng
- National Health Commission (NHC) Key Laboratory of Transplant Engineering and Immunology, Regenerative Medicine Research Center, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Jingping Liu
- National Health Commission (NHC) Key Laboratory of Transplant Engineering and Immunology, Regenerative Medicine Research Center, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
- *Correspondence: Jingping Liu,
| |
Collapse
|
32
|
Bai YT, Zhang XQ, Chen XJ, Zhou G. Nanomedicines in oral cancer: inspiration comes from extracellular vesicles and biomimetic nanoparticles. Nanomedicine (Lond) 2022; 17:1761-1778. [PMID: 36647844 DOI: 10.2217/nnm-2022-0142] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Oral cancer is a common life-threatening malignancy having high mortality and morbidity rates. During the treatment process, individuals unavoidably experience severe side effects. It is essential to develop safer and more effective strategies. Currently, extracellular vesicles (EVs) and biomimetic nanoparticles are nanomedicines with long-term blood circulation and lower off-target toxicity that orchestrate immune responses and accumulate specifically in tumor sites. EVs create a synergetic effect by encapsulating drugs and collaborating with naturally loaded elements in the EVs. Biomimetic nanoparticles retain the characteristic features of the synthetic nanocarriers and inherit the intrinsic cell membrane functionalities. This review outlines the properties, applications, challenges, pros and cons of EVs and biomimetic nanoparticles, providing novel perspectives on oral cancer.
Collapse
Affiliation(s)
- Yu-Ting Bai
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Xue-Qiong Zhang
- School of Chemistry, Chemical Engineering & Life Sciences, Wuhan University of Technology, Wuhan, 430070, China
| | - Xiao-Jie Chen
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China.,Department of Oral Medicine, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Gang Zhou
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China.,Department of Oral Medicine, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| |
Collapse
|
33
|
Yoshitake J, Azami M, Sei H, Onoshima D, Takahashi K, Hirayama A, Uchida K, Baba Y, Shibata T. Rapid Isolation of Extracellular Vesicles Using a Hydrophilic Porous Silica Gel-Based Size-Exclusion Chromatography Column. Anal Chem 2022; 94:13676-13681. [PMID: 36166570 PMCID: PMC9558085 DOI: 10.1021/acs.analchem.2c01053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
![]()
Extracellular vesicles
(EVs) are nanoscale lipid bilayer vesicles
released by almost all cell types and can be found in biological fluids,
such as blood and urine. EVs play an important role in various physiological
and pathological processes via cell–cell communication, highlighting
their potential applications as diagnostic markers for diseases and
therapeutic drug delivery carriers. Although various methods have
been developed for the isolation of EVs from biological fluids, most
of them exhibit major limitations, including low purity, long processing
times, and high cost. In this study, we developed a size-exclusion
chromatography (SEC) column device using hydrophilic porous silica
gel (PSG). Owing to the resistance to pressure of the device, a rapid
system for EV isolation was developed by connecting it to a flash
liquid chromatography system furnished with a UV detector and a fraction
collector. This system can be used for the real-time monitoring of
eluted EVs by UV absorption without further analysis and separation
of high-purity EVs from urine samples with high durability, reusability,
and reproducibility. In addition, there were no significant differences
between the PSG column- and conventional SEC column-isolated EVs in
the proteome profiles and cellular uptake activities, suggesting the
good quality of the EVs isolated by the PSG column. These findings
suggest that the PSG column device offers an effective and rapid method
for the isolation of intact EVs from biological fluids.
Collapse
Affiliation(s)
- Jun Yoshitake
- Institute of Nano-Life-Systems, Institutes of Innovation for Future Society, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8603, Japan
| | - Mayuko Azami
- Graduate School of Bioagricultural Sciences, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8601, Japan
| | - Haruka Sei
- Graduate School of Bioagricultural Sciences, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8601, Japan
| | - Daisuke Onoshima
- Institute of Nano-Life-Systems, Institutes of Innovation for Future Society, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8603, Japan
| | - Kumiko Takahashi
- Materials Integration Laboratories, AGC Inc., 1-1, Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan
| | - Akiyoshi Hirayama
- Institute of Nano-Life-Systems, Institutes of Innovation for Future Society, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8603, Japan.,Institute for Advanced Biosciences, Keio University, 246-2 Mizukami, Kakuganji, Tsuruoka, Yamagata 997-0052, Japan
| | - Koji Uchida
- Graduate School of Agricultural and Life Sciences, The University of Tokyo, Yayoi 1-1-1, Bunkyo-ku, Tokyo 113-8657, Japan.,Japan Agency for Medical Research and Development, CREST, 1-7-1 Otemachi, Chiyoda-ku, Tokyo 100-0004, Japan
| | - Yoshinobu Baba
- Institute of Nano-Life-Systems, Institutes of Innovation for Future Society, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8603, Japan.,Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8603, Japan.,Institute of Quantum Life Science, National Institutes for Quantum and Radiological Science and Technology, Anagawa 4-9-1, Inage-ku, Chiba 263-8555, Japan.,College of Pharmacy, Kaohsiung Medical University, 100, Shin-Chuan 1st Rd., Kaohsiung 807, Taiwan
| | - Takahiro Shibata
- Institute of Nano-Life-Systems, Institutes of Innovation for Future Society, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8603, Japan.,Graduate School of Bioagricultural Sciences, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8601, Japan.,Institute for Glyco-Core Research (iGCORE), Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8601, Japan
| |
Collapse
|
34
|
Shen X, Song J, Kawakami K, Ariga K. Molecule-to-Material-to-Bio Nanoarchitectonics with Biomedical Fullerene Nanoparticles. MATERIALS (BASEL, SWITZERLAND) 2022; 15:5404. [PMID: 35955337 PMCID: PMC9369991 DOI: 10.3390/ma15155404] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 07/28/2022] [Accepted: 08/02/2022] [Indexed: 06/15/2023]
Abstract
Nanoarchitectonics integrates nanotechnology with various other fields, with the goal of creating functional material systems from nanoscale units such as atoms, molecules, and nanomaterials. The concept bears strong similarities to the processes and functions seen in biological systems. Therefore, it is natural for materials designed through nanoarchitectonics to truly shine in bio-related applications. In this review, we present an overview of recent work exemplifying how nanoarchitectonics relates to biology and how it is being applied in biomedical research. First, we present nanoscale interactions being studied in basic biology and how they parallel nanoarchitectonics concepts. Then, we overview the state-of-the-art in biomedical applications pursuant to the nanoarchitectonics framework. On this basis, we take a deep dive into a particular building-block material frequently seen in nanoarchitectonics approaches: fullerene. We take a closer look at recent research on fullerene nanoparticles, paying special attention to biomedical applications in biosensing, gene delivery, and radical scavenging. With these subjects, we aim to illustrate the power of nanomaterials and biomimetic nanoarchitectonics when applied to bio-related applications, and we offer some considerations for future perspectives.
Collapse
Affiliation(s)
- Xuechen Shen
- Graduate School of Frontier Sciences, The University of Tokyo, 5-1-5 Kashiwanoha, Kashiwa 277-8561, Chiba, Japan
| | - Jingwen Song
- Research Center for Functional Materials, National Institute for Materials Science (NIMS), 1-1 Namiki, Tsukuba 305-0044, Ibaraki, Japan
| | - Kohsaku Kawakami
- Research Center for Functional Materials, National Institute for Materials Science (NIMS), 1-1 Namiki, Tsukuba 305-0044, Ibaraki, Japan
- Graduate School of Pure and Applied Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba 305-8577, Ibaraki, Japan
| | - Katsuhiko Ariga
- Graduate School of Frontier Sciences, The University of Tokyo, 5-1-5 Kashiwanoha, Kashiwa 277-8561, Chiba, Japan
- WPI Research Center for Materials Nanoarchitectonics (MANA), National Institute for Materials Science (NIMS), 1-1 Namiki, Tsukuba 305-0044, Ibaraki, Japan
| |
Collapse
|
35
|
Exosomes as Crucial Players in Pathogenesis of Systemic Lupus Erythematosus. J Immunol Res 2022; 2022:8286498. [PMID: 35910853 PMCID: PMC9328965 DOI: 10.1155/2022/8286498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 06/29/2022] [Indexed: 11/18/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is a systemic autoimmune disease that affects multiple systems. Its clinical manifestation varies across patients, from skin mucosa to multiorgan damage to severe central nervous system involvement. The exosome has been shown to play an important role in the pathogenesis of autoimmune diseases, including SLE. We review the recent knowledge of exosomes, including their biology, functions, mechanism, and standardized extraction and purification methods in SLE, to highlight potential therapeutic targets for SLE.
Collapse
|
36
|
Prasad R, Conde J. Bioinspired soft nanovesicles for site-selective cancer imaging and targeted therapies. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2022; 14:e1792. [PMID: 35318815 DOI: 10.1002/wnan.1792] [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: 11/05/2021] [Revised: 02/17/2022] [Accepted: 03/03/2022] [Indexed: 06/14/2023]
Abstract
Cell-to-cell communication within the heterogeneous solid tumor environment plays a significant role in the uncontrolled metastasis of cancer. To inhibit the metastasis and growth of cancer cells, various chemically designed and biologically derived nanosized biomaterials have been applied for targeted cancer therapeutics applications. Over the years, bioinspired soft nanovesicles have gained tremendous attention for targeted cancer therapeutics due to their easy binding with tumor microenvironment, natural targeting ability, bio-responsive nature, better biocompatibility, high cargo capacity for multiple therapeutics agents, and long circulation time. These cell-derived nanovesicles guard their loaded cargo molecules from immune clearance and make them site-selective to cancer cells due to their natural binding and delivery abilities. Furthermore, bioinspired soft nanovesicles prevent cell-to-cell communication and secretion of cancer cell markers by delivering the therapeutics agents predominantly. Cell-derived vesicles, namely, exosomes, extracellular vesicles, and so forth have been recognized as versatile carriers for therapeutic biomolecules. However, low product yield, poor reproducibility, and uncontrolled particle size distribution have remained as major challenges of these soft nanovesicles. Furthermore, the surface biomarkers and molecular contents of these vesicles change with respect to the stage of disease and types. Here in this review, we have discussed numerous examples of bioinspired soft vesicles for targeted imaging and cancer therapeutic applications with their advantages and limitations. Importance of bioengineered soft nanovesicles for localized therapies with their clinical relevance has also been addressed in this article. Overall, cell-derived nanovesicles could be considered as clinically relevant platforms for cancer therapeutics. This article is categorized under: Biology-Inspired Nanomaterials > Nucleic Acid-Based Structures Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease.
Collapse
Affiliation(s)
- Rajendra Prasad
- NOVA Medical School, Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Lisbon, Portugal
| | - João Conde
- NOVA Medical School, Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Lisbon, Portugal
- Centre for Toxicogenomics and Human Health, Genetics, Oncology and Human Toxicology, NOVA Medical School, Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Lisbon, Portugal
| |
Collapse
|
37
|
Nishat ZS, Hossain T, Islam MN, Phan HP, Wahab MA, Moni MA, Salomon C, Amin MA, Sina AAI, Hossain MSA, Kaneti YV, Yamauchi Y, Masud MK. Hydrogel Nanoarchitectonics: An Evolving Paradigm for Ultrasensitive Biosensing. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2107571. [PMID: 35620959 DOI: 10.1002/smll.202107571] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 03/02/2022] [Indexed: 06/15/2023]
Abstract
The integration of nanoarchitectonics and hydrogel into conventional biosensing platforms offers the opportunities to design physically and chemically controlled and optimized soft structures with superior biocompatibility, better immobilization of biomolecules, and specific and sensitive biosensor design. The physical and chemical properties of 3D hydrogel structures can be modified by integrating with nanostructures. Such modifications can enhance their responsiveness to mechanical, optical, thermal, magnetic, and electric stimuli, which in turn can enhance the practicality of biosensors in clinical settings. This review describes the synthesis and kinetics of gel networks and exploitation of nanostructure-integrated hydrogels in biosensing. With an emphasis on different integration strategies of hydrogel with nanostructures, this review highlights the importance of hydrogel nanostructures as one of the most favorable candidates for developing ultrasensitive biosensors. Moreover, hydrogel nanoarchitectonics are also portrayed as a promising candidate for fabricating next-generation robust biosensors.
Collapse
Affiliation(s)
- Zakia Sultana Nishat
- Department of Biochemistry and Molecular Biology, School of Life Sciences, Shahjalal University of Science and Technology, Sylhet, 3114, Bangladesh
| | - Tanvir Hossain
- Department of Biochemistry and Molecular Biology, School of Life Sciences, Shahjalal University of Science and Technology, Sylhet, 3114, Bangladesh
- Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Md Nazmul Islam
- School of Health and Life Sciences, Teesside University, Tees Valley, Middlesbrough, TS1 3BA, UK
| | - Hoang-Phuong Phan
- Queensland Micro and Nanotechnology Centre, Griffith University, Nathan, QLD, 4111, Australia
| | - Md A Wahab
- Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Mohammad Ali Moni
- School of Health and Rehabilitation Sciences, Faculty of Health and Behavioural Sciences, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Carlos Salomon
- Exosome Biology Laboratory, Centre for Clinical Diagnostics, University of Queensland Centre for Clinical Research, Royal Brisbane and Women's Hospital Faculty of Medicine, The University of Queensland, Herston, Brisbane City, QLD, 4029, Australia
- Departamento de Investigación, Postgrado y Educación Continua (DIPEC), Facultad de Ciencias de la Salud, Universidad del Alba, Santiago, 8320000, Chile
| | - Mohammed A Amin
- Department of Chemistry, College of Science, Taif University, P. O. Box 11099, Taif, 21944, Saudi Arabia
| | - Abu Ali Ibn Sina
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard University, Boston, MA, 02115, USA
| | - Md Shahriar A Hossain
- Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, Brisbane, QLD, 4072, Australia
- School of Mechanical and Mining Engineering, Faculty of Engineering, Architecture and Information Technology (EAIT), The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Yusuf Valentino Kaneti
- Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Yusuke Yamauchi
- Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, Brisbane, QLD, 4072, Australia
- School of Chemical Engineering, Faculty of Engineering, Architecture and Information Technology (EAIT), The University of Queensland, Brisbane, QLD, 4072, Australia
- JST-ERATO Yamauchi Materials Space-Tectonics Project and International Center for Materials Nanoarchitectonics (WPI-MANA), National Institute for Materials Science, Tsukuba, Ibaraki, 305-0044, Japan
| | - Mostafa Kamal Masud
- Department of Biochemistry and Molecular Biology, School of Life Sciences, Shahjalal University of Science and Technology, Sylhet, 3114, Bangladesh
- Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, Brisbane, QLD, 4072, Australia
- JST-ERATO Yamauchi Materials Space-Tectonics Project and International Center for Materials Nanoarchitectonics (WPI-MANA), National Institute for Materials Science, Tsukuba, Ibaraki, 305-0044, Japan
| |
Collapse
|
38
|
Fan Z, Jiang C, Wang Y, Wang K, Marsh J, Zhang D, Chen X, Nie L. Engineered extracellular vesicles as intelligent nanosystems for next-generation nanomedicine. NANOSCALE HORIZONS 2022; 7:682-714. [PMID: 35662310 DOI: 10.1039/d2nh00070a] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Extracellular vesicles (EVs), as natural carriers of bioactive cargo, have a unique micro/nanostructure, bioactive composition, and characteristic morphology, as well as fascinating physical, chemical and biochemical features, which have shown promising application in the treatment of a wide range of diseases. However, native EVs have limitations such as lack of or inefficient cell targeting, on-demand delivery, and therapeutic feedback. Recently, EVs have been engineered to contain an intelligent core, enabling them to (i) actively target sites of disease, (ii) respond to endogenous and/or exogenous signals, and (iii) provide treatment feedback for optimal function in the host. These advances pave the way for next-generation nanomedicine and offer promise for a revolution in drug delivery. Here, we summarise recent research on intelligent EVs and discuss the use of "intelligent core" based EV systems for the treatment of disease. We provide a critique about the construction and properties of intelligent EVs, and challenges in their commercialization. We compare the therapeutic potential of intelligent EVs to traditional nanomedicine and highlight key advantages for their clinical application. Collectively, this review aims to provide a new insight into the design of next-generation EV-based theranostic platforms for disease treatment.
Collapse
Affiliation(s)
- Zhijin Fan
- School of Medicine, South China University of Technology, Guangzhou 510006, P. R. China.
- Research Center of Medical Sciences, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, P. R. China
| | - Cheng Jiang
- School of Life and Health Sciences, The Chinese University of Hong Kong, Shenzhen 518172, China
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Yichao Wang
- Department of Clinical Laboratory Medicine, Tai Zhou Central Hospital (Taizhou University Hospital), Taizhou 318000, P. R. China
| | - Kaiyuan Wang
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, P. R. China
| | - Jade Marsh
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Da Zhang
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, P. R. China.
| | - Xin Chen
- School of Chemical Engineering and Technology, Shaanxi Key Laboratory of Energy Chemical Process Intensification, Institute of Polymer Science in Chemical Engineering, Xi'an Jiao Tong University, Xi'an 710049, P. R. China.
| | - Liming Nie
- Research Center of Medical Sciences, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, P. R. China
- School of Medicine, South China University of Technology, Guangzhou 510006, P. R. China.
| |
Collapse
|
39
|
Liu X, Zhang G, Yu T, He J, Liu J, Chai X, Zhao G, Yin D, Zhang C. Exosomes deliver lncRNA DARS-AS1 siRNA to inhibit chronic unpredictable mild stress-induced TNBC metastasis. Cancer Lett 2022; 543:215781. [PMID: 35688263 DOI: 10.1016/j.canlet.2022.215781] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 05/09/2022] [Accepted: 06/01/2022] [Indexed: 11/19/2022]
Abstract
Triple-negative breast cancer (TNBC) is a rapidly recurring and highly metastatic malignancy with high heterogeneity and chemoradiotherapy resistance. Chronic unpredictable mild stress (CUMS) can induce the occurrence of tumors and enhance lymphatic infiltration and distant metastasis through direct interaction with the sympathetic nervous system; however, its relevance in TNBC is yet to be clarified. In this study, DARS-AS1, a newly reported CUMS-responsive lncRNA, was found to be enriched in TNBC clinical tumors and cells and positively correlated with late clinical stage in patients with TNBC. DARS-AS1 overexpression significantly enhanced the migration and invasion of TNBC tumors by inhibiting miR-129-2-3p and upregulated CDK1 to activate the NF-κB/STAT3 signaling pathway both in vitro and in vivo. Treatment with DARS-AS1 siRNA-loaded exosomes (EXOs) substantially slowed CUMS-induced TNBC cell growth and liver metastasis. Therefore, DARS-AS1 represents a potential therapeutic target for metastatic TNBC, and EXOs may serve as siRNA delivery carriers in clinical therapy.
Collapse
Affiliation(s)
- Xinli Liu
- Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Ge Zhang
- Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Tongyao Yu
- Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Jinliang He
- Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Jie Liu
- Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Xiaoxia Chai
- Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Gang Zhao
- The First Hospital of Jilin University, Changchun, 130021, China.
| | - Dachuan Yin
- Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, China.
| | - Chenyan Zhang
- Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, China.
| |
Collapse
|
40
|
Liu H, Liang J, Ye X, Huang M, Ma L, Xie X, Liu D, Cao H, Simal-Gandara J, Rengasamy KRR, Wang Q, Xiao G, Xiao J. The potential role of extracellular vesicles in bioactive compound-based therapy: A review of recent developments. Crit Rev Food Sci Nutr 2022; 63:10959-10973. [PMID: 35648042 DOI: 10.1080/10408398.2022.2081667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Recent studies have explored the field of extracellular vesicles (EVs), driving an increasing interest in their application to human health. EVs have unique physicochemical traits to participate in intercellular communication, thus fostering the idea of using EVs to yield synergistic, preventive, and therapeutic effects. Many reports have shown that EVs contain natural bioactive compounds, such as lipids, proteins, RNA, and other active components that regulate biological processes, thereby contributing to human health. Therefore, in this review, we comprehensively elucidate various facets of the relationship between EVs and bioactive compounds that modulate EVs contents, including RNAs and proteins, discussing different forms of biological regulation. The use of EVs for cargo-loading bioactive compounds to exert biological functions and methods to load bioactive compounds into EVs are also discussed. This review highlighted the effect of EV-delivered bioactive compounds on several therapeutic mechanisms and applications, providing new insight into nutrition and pharmacology.
Collapse
Affiliation(s)
- Huifan Liu
- College of Light Industry and Food, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Lingnan Specialty Food Science and Technology, Guangzhou, Guangdong, China
| | - Jiaxi Liang
- College of Light Industry and Food, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong, China
| | - Xia Ye
- College of Light Industry and Food, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong, China
| | - Miaoru Huang
- College of Light Industry and Food, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong, China
| | - Lukai Ma
- College of Light Industry and Food, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Lingnan Specialty Food Science and Technology, Guangzhou, Guangdong, China
| | - Xi Xie
- College of Light Industry and Food, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Lingnan Specialty Food Science and Technology, Guangzhou, Guangdong, China
| | - Dongjie Liu
- College of Light Industry and Food, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Lingnan Specialty Food Science and Technology, Guangzhou, Guangdong, China
| | - Hui Cao
- Universidade de Vigo, Nutrition and Bromatology Group, Department of Analytical and Food Chemistry, Faculty of Sciences, Ourense, Spain
| | - Jesus Simal-Gandara
- Universidade de Vigo, Nutrition and Bromatology Group, Department of Analytical and Food Chemistry, Faculty of Sciences, Ourense, Spain
| | - Kannan R R Rengasamy
- Center for Transdisciplinary Research, Department of Pharmacology, Saveetha Dental College, Saveetha Institute of Medical and Technical Science, Saveetha University, Chennai, India
| | - Qin Wang
- College of Light Industry and Food, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Lingnan Specialty Food Science and Technology, Guangzhou, Guangdong, China
| | - Gengsheng Xiao
- College of Light Industry and Food, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Lingnan Specialty Food Science and Technology, Guangzhou, Guangdong, China
| | - Jianbo Xiao
- Universidade de Vigo, Nutrition and Bromatology Group, Department of Analytical and Food Chemistry, Faculty of Sciences, Ourense, Spain
- International Research Center for Food Nutrition and Safety, Jiangsu University, Zhenjiang, China
| |
Collapse
|
41
|
Ariga K. Mechano-Nanoarchitectonics: Design and Function. SMALL METHODS 2022; 6:e2101577. [PMID: 35352500 DOI: 10.1002/smtd.202101577] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Revised: 03/12/2022] [Indexed: 05/27/2023]
Abstract
Mechanical stimuli have rather ambiguous and less-specific features among various physical stimuli, but most materials exhibit a certain level of responses upon mechanical inputs. Unexplored sciences remain in mechanical responding systems as one of the frontiers of materials science. Nanoarchitectonics approaches for mechanically responding materials are discussed as mechano-nanoarchitectonics in this review article. Recent approaches on molecular and materials systems with mechanical response capabilities are first exemplified with two viewpoints: i) mechanical control of supramolecular assemblies and materials and ii) mechanical control and evaluation of atom/molecular level structures. In the following sections, special attentions on interfacial environments for mechano-nanoarchitectonics are emphasized. The section entitled iii) Mechanical Control of Molecular System at Dynamic Interface describes coupling of macroscopic mechanical forces and molecular-level phenomena. Delicate mechanical forces can be applied to functional molecules embedded at the air-water interface where operation of molecular machines and tuning of molecular receptors upon macroscopic mechanical actions are discussed. Finally, the important role of the interfacial media are further extended to the control of living cells as described in the section entitled iv) Mechanical Control of Biosystems. Pioneering approaches on cell fate regulations at liquid-liquid interfaces are discussed in addition to well-known mechanobiology.
Collapse
Affiliation(s)
- Katsuhiko Ariga
- World Premier International (WPI) Research Center for Materials Nanoarchitectonics (MANA), National Institute for Materials Science (NIMS), 1-1 Namiki, Tsukuba, 305-0044, Japan
- Department of Advanced Materials Science, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Chiba, 277-8561, Japan
| |
Collapse
|
42
|
Liu H, Geng Z, Su J. Engineered mammalian and bacterial extracellular vesicles as promising nanocarriers for targeted therapy. EXTRACELLULAR VESICLES AND CIRCULATING NUCLEIC ACIDS 2022; 3:63-86. [PMID: 39698442 PMCID: PMC11648430 DOI: 10.20517/evcna.2022.04] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 03/21/2022] [Accepted: 04/07/2022] [Indexed: 12/20/2024]
Abstract
Extracellular vesicles (EVs), which are nanocarriers with phospholipid bilayer structures released by most cells, play a key role in regulating physiological and pathological processes. EVs have been investigated due to their loading capacity, low toxicity, immunogenicity, and biofunctions. Although EVs have shown good potential as therapeutic vehicles, natural EVs have a poor targeting ability, which substantially reduces the therapeutic effect. Through the addition of a targeting unit into the membrane surface of EVs or inside EVs by engineering technology, the therapeutic agent can accumulate in specific cells and tissues. Here, we focus on mammalian EVs (MEVs) and bacterial EVs (BEVs), which are the two most common types of EVs in the biomedical field. In this review, we describe engineered MEVs and BEVs as promising nanocarriers for targeted therapy and summarize the biogenesis, isolation, and characterization of MEVs and BEVs. We then describe engineering techniques for enhancement of the targeting ability of EVs. Moreover, we focus on the applications of engineered MEVs and BEVs in targeted therapy, including the treatment of cancer and brain and bone disease. We believe that this review will help improve the understanding of engineered MEVs and BEVs, thereby promoting their application and clinical translation.
Collapse
Affiliation(s)
| | | | - Jiacan Su
- Correspondence to: Prof. Jiacan Su, Institute of Translational Medicine, Shanghai University, Shanghai 200444, China. E-mail:
| |
Collapse
|
43
|
Shen X, Song J, Sevencan C, Leong DT, Ariga K. Bio-interactive nanoarchitectonics with two-dimensional materials and environments. SCIENCE AND TECHNOLOGY OF ADVANCED MATERIALS 2022; 23:199-224. [PMID: 35370475 PMCID: PMC8973389 DOI: 10.1080/14686996.2022.2054666] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 03/12/2022] [Accepted: 03/15/2022] [Indexed: 05/19/2023]
Abstract
Like the proposal of nanotechnology by Richard Feynman, the nanoarchitectonics concept was initially proposed by Masakazu Aono. The nanoarchitectonics strategy conceptually fuses nanotechnology with other research fields including organic chemistry, supramolecular chemistry, micro/nanofabrication, materials science, and bio-related sciences, and aims to produce functional materials from nanoscale components. In this review article, bio-interactive nanoarchitectonics and two-dimensional materials and environments are discussed as a selected topic. The account gives general examples of nanoarchitectonics of two-dimensional materials for energy storage, catalysis, and biomedical applications, followed by explanations of bio-related applications with two-dimensional materials such as two-dimensional biomimetic nanosheets, fullerene nanosheets, and two-dimensional assemblies of one-dimensional fullerene nanowhiskers (FNWs). The discussion on bio-interactive nanoarchitectonics in two-dimensional environments further extends to liquid-liquid interfaces such as fluorocarbon-medium interfaces and viscous liquid interfaces as new frontiers of two-dimensional environments for bio-related applications. Controlling differentiation of stem cells at fluidic liquid interfaces is also discussed. Finally, a conclusive section briefly summarizes features of bio-interactive nanoarchitectonics with two-dimensional materials and environments and discusses possible future perspectives.
Collapse
Affiliation(s)
- Xuechen Shen
- Graduate School of Frontier Sciences, The University of Tokyo, Chiba, Japan
- WPI Research Center for Materials Nanoarchitectonics (MANA), National Institute for Materials Science (NIMS), Ibaraki, Japan
| | - Jingwen Song
- Graduate School of Frontier Sciences, The University of Tokyo, Chiba, Japan
- WPI Research Center for Materials Nanoarchitectonics (MANA), National Institute for Materials Science (NIMS), Ibaraki, Japan
| | - Cansu Sevencan
- Department of Chemical and Biomolecular Engineering, National University of Singapore, Singapore, Singapore
| | - David Tai Leong
- Department of Chemical and Biomolecular Engineering, National University of Singapore, Singapore, Singapore
| | - Katsuhiko Ariga
- Graduate School of Frontier Sciences, The University of Tokyo, Chiba, Japan
- WPI Research Center for Materials Nanoarchitectonics (MANA), National Institute for Materials Science (NIMS), Ibaraki, Japan
| |
Collapse
|
44
|
Paisrisarn P, Yasui T, Zhu Z, Klamchuen A, Kasamechonchung P, Wutikhun T, Yordsri V, Baba Y. Tailoring ZnO nanowire crystallinity and morphology for label-free capturing of extracellular vesicles. NANOSCALE 2022; 14:4484-4494. [PMID: 35234770 DOI: 10.1039/d1nr07237d] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Zinc oxide (ZnO) nanowires have shown their potential in isolation of cancer-related biomolecules such as extracellular vesicles (EVs), RNAs, and DNAs for early diagnosis and therapeutic development of diseases. Since the function of inorganic nanowires changes depending on their morphology, previous studies have established strategies to control the morphology and have demonstrated attainment of improved properties for gas and organic compound detection, and for dye-sensitized solar cells and photoelectric conversion performance. Nevertheless, crystallinity and morphology of ZnO nanowires for capturing EVs, an important biomarker of cancer, have not yet been discussed. Here, we fabricated ZnO nanowires with different crystallinities and morphologies using an ammonia-assisted hydrothermal method, and we comprehensively analyzed the crystalline nature and oriented growth of the synthesized nanowires by X-ray diffraction and selected area electron diffraction using high resolution transmission electron microscopy. In evaluating the performance of label-free EV capture in a microfluidic device platform, we found both the crystallinity and morphology of ZnO nanowires affected EV capture efficiency. In particular, the zinc blende phase was identified as important for crystallinity, while increasing the nanowire density in the array was important for morphology to improve EV capture performance. These results highlighted that the key physicochemical properties of the ZnO nanowires were related to the EV capture performance.
Collapse
Affiliation(s)
- Piyawan Paisrisarn
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8603, Japan.
| | - Takao Yasui
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8603, Japan.
- Japan Science and Technology Agency (JST), PRESTO, 4-1-8 Honcho, Kawaguchi, Saitama 332-0012, Japan
- Institute of Nano-Life-Systems, Institutes of Innovation for Future Society, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8603, Japan
| | - Zetao Zhu
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8603, Japan.
- Institute of Nano-Life-Systems, Institutes of Innovation for Future Society, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8603, Japan
| | - Annop Klamchuen
- National Nanotechnology Center (NANOTEC), National Science and Technology Development Agency (NSTDA), Pathum Thani 12120, Thailand
| | - Panita Kasamechonchung
- National Nanotechnology Center (NANOTEC), National Science and Technology Development Agency (NSTDA), Pathum Thani 12120, Thailand
| | - Tuksadon Wutikhun
- National Nanotechnology Center (NANOTEC), National Science and Technology Development Agency (NSTDA), Pathum Thani 12120, Thailand
| | - Visittapong Yordsri
- National Metal and Materials Technology Center (MTEC), National Science and Technology Development Agency (NSTDA), Pathum Thani 12120, Thailand
| | - Yoshinobu Baba
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8603, Japan.
- Institute of Nano-Life-Systems, Institutes of Innovation for Future Society, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8603, Japan
- Institute of Quantum Life Science, National Institutes for Quantum and Radiological Science and Technology, Anagawa 4-9-1, Inage-ku, Chiba 263-8555, Japan
| |
Collapse
|
45
|
Exosomes from Bone Marrow Mesenchymal Stem Cells with Overexpressed Nrf2 Inhibit Cardiac Fibrosis in Rats with Atrial Fibrillation. Cardiovasc Ther 2022; 2022:2687807. [PMID: 35360547 PMCID: PMC8941574 DOI: 10.1155/2022/2687807] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 02/26/2022] [Accepted: 03/03/2022] [Indexed: 12/18/2022] Open
Abstract
Background Even though nuclear factor-erythroid 2-related factor 2 (Nrf2) signaling has been associated with the pathogenesis of multiple heart conditions, data on roles of Nrf2 within atrial fibrillation (AF) still remain scant. The present investigation had the aim of analyzing Nrf2-overexpressing role/s upon bone mesenchymal stem cell- (BMSC-) derived exosomes in rats with AF. Methods Exosomes were collected from control or Nrf2 lentivirus-transduced BMSCs and then injected into rats with AF through the tail vein. AF duration was observed using electrocardiography. Immunohistochemical staining was then employed for assessing Nrf2, HO-1, α-SMA, collagen I, or TGF-β1 expression profiles within atrial myocardium tissues. Conversely, Masson staining was utilized to evaluate atrial fibrosis whereas apoptosis within myocardia was evaluated through TUNEL assays. In addition, TNF-α, IL-1β, IL-4, or IL-10 serum expression was assessed through ELISA. Results Results of the current study showed significant downregulation of Nrf2/HO-1 within AF rat myocardia. It was found that injection of the control or Lv-Nrf2 exosomes significantly alleviated and lowered AF timespans together with reducing cardiomyocyte apoptosis. Moreover, injection of Lv-Nrf2 exosomes essentially lowered AF-driven atrial fibrosis and also inhibited inflammatory responses in the rats with AF. Conclusion Delivery of BMSC-derived exosomes using overexpressed Nrf2 inhibited AF-induced arrhythmias, myocardial fibrosis, apoptosis, and inflammation via Nrf2/HO-1 pathway triggering.
Collapse
|
46
|
Li F, Wu J, Li D, Hao L, Li Y, Yi D, Yeung KWK, Chen D, Lu WW, Pan H, Wong TM, Zhao X. Engineering stem cells to produce exosomes with enhanced bone regeneration effects: an alternative strategy for gene therapy. J Nanobiotechnology 2022; 20:135. [PMID: 35292020 PMCID: PMC8922796 DOI: 10.1186/s12951-022-01347-3] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Accepted: 03/02/2022] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Exosomes derived from stem cells have been widely studied for promoting regeneration and reconstruction of multiple tissues as "cell-free" therapies. However, the applications of exosomes have been hindered by limited sources and insufficient therapeutic potency. RESULTS In this study, a stem cell-mediated gene therapy strategy is developed in which mediator mesenchymal stem cells are genetically engineered by bone morphogenetic protein-2 gene to produce exosomes (MSC-BMP2-Exo) with enhanced bone regeneration potency. This effect is attributed to the synergistic effect of the content derived from MSCs and the up-regulated BMP2 gene expression. The MSC-BMP2-Exo also present homing ability to the injured site. The toxic effect of genetical transfection vehicles is borne by mediator MSCs, while the produced exosomes exhibit excellent biocompatibility. In addition, by plasmid tracking, it is interesting to find a portion of plasmid DNA can be encapsulated by exosomes and delivered to recipient cells. CONCLUSIONS In this strategy, engineered MSCs function as cellular factories, which effectively produce exosomes with designed and enhanced therapeutic effects. The accelerating effect in bone healing and the good biocompatibility suggest the potential clinical application of this strategy.
Collapse
Affiliation(s)
- Feiyang Li
- Research Center for Human Tissue and Organs Degeneration, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Jun Wu
- Shenzhen Key Laboratory for Innovative Technology in Orthopaedic Trauma, The University of Hong Kong-Shenzhen Hospital, Shenzhen, 518053, China
- Department of Orthopaedics and Traumatology, The University of Hong Kong, Hong Kong, 999077, China
| | - Daiye Li
- Shenzhen Key Laboratory for Innovative Technology in Orthopaedic Trauma, The University of Hong Kong-Shenzhen Hospital, Shenzhen, 518053, China
- Department of Orthopaedics and Traumatology, The University of Hong Kong, Hong Kong, 999077, China
| | - Liuzhi Hao
- Research Center for Human Tissue and Organs Degeneration, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yanqun Li
- Research Center for Human Tissue and Organs Degeneration, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Dan Yi
- Research Center for Human Tissue and Organs Degeneration, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Kelvin W K Yeung
- Shenzhen Key Laboratory for Innovative Technology in Orthopaedic Trauma, The University of Hong Kong-Shenzhen Hospital, Shenzhen, 518053, China
- Department of Orthopaedics and Traumatology, The University of Hong Kong, Hong Kong, 999077, China
| | - Di Chen
- Research Center for Human Tissue and Organs Degeneration, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - William W Lu
- Research Center for Human Tissue and Organs Degeneration, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
- Department of Orthopaedics and Traumatology, The University of Hong Kong, Hong Kong, 999077, China
| | - Haobo Pan
- Research Center for Human Tissue and Organs Degeneration, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Tak Man Wong
- Shenzhen Key Laboratory for Innovative Technology in Orthopaedic Trauma, The University of Hong Kong-Shenzhen Hospital, Shenzhen, 518053, China.
- Department of Orthopaedics and Traumatology, The University of Hong Kong, Hong Kong, 999077, China.
| | - Xiaoli Zhao
- Research Center for Human Tissue and Organs Degeneration, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
47
|
Recent Advances in the Application of Mesenchymal Stem Cell-Derived Exosomes for Cardiovascular and Neurodegenerative Disease Therapies. Pharmaceutics 2022; 14:pharmaceutics14030618. [PMID: 35335993 PMCID: PMC8949563 DOI: 10.3390/pharmaceutics14030618] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Revised: 03/02/2022] [Accepted: 03/09/2022] [Indexed: 12/17/2022] Open
Abstract
Exosomes are naturally occurring nanoscale vesicles that are released and received by almost all cells in the body. Exosomes can be transferred between cells and contain various molecular constitutes closely related to their origin and function, including proteins, lipids, and RNAs. The importance of exosomes in cellular communication makes them important vectors for delivering a variety of drugs throughout the body. Exosomes are ubiquitous in the circulatory system and can reach the site of injury or disease through a variety of biological barriers. Due to its unique structure and rich inclusions, it can be used for the diagnosis and treatment of diseases. Mesenchymal stem-cell-derived exosomes (MSCs-Exo) inherit the physiological functions of MSCs, including repairing and regenerating tissues, suppressing inflammatory responses, and regulating the body’s immunity; therefore, MSCs-Exo can be used as a natural drug delivery carrier with therapeutic effects, and has been increasingly used in the treatment of cardiovascular diseases and neurodegenerative diseases. Here, we summarize the research progress of MSCs-Exo as drug delivery vectors and their application for various drug deliveries, providing ideas and references for the study of MSCs-Exo in recent years.
Collapse
|
48
|
Bhattacharya S, Patel R, Joshi A. The Most Recent Discoveries in Heterocyclic Nanoformulations for Targeted Anticancer Therapy. Mini Rev Med Chem 2022; 22:1735-1751. [PMID: 35114919 DOI: 10.2174/138955752203220202164839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 09/02/2021] [Accepted: 11/15/2021] [Indexed: 11/22/2022]
Abstract
Every day, new cases of cancer patients whose recovery is delayed by multidrug resistance and chemotherapy side effects are identified, which severely limit treatment options. One of the most recent advances in nanotechnology is the effective usage of nanotechnology as drug carriers for cancer therapy. As a consequence, heterocyclic nanocarriers were put into practice to see whether they could have a better cure with positive results. The potential of a therapeutic agent to meet its desired goal is vital to its success in treating any disease. Heterocyclic moieties are molecules that have a wide variety of chemically therapeutic functions as well as a significant biological activity profile. Heterocyclic nano formulations play an important role in cell physiology and as possible arbitrators for typical biological reactions, making them valuable in cancer research. As a result, experts are working with heterocyclic nanoformulations to discover alternative approaches to treat cancer. Due to their unique physicochemical properties, heterocyclic compounds are real cornerstones in medicinal chemistry and promising compounds for the future drug delivery system. This review briefly explores the therapeutic relevance of heterocyclic compounds in cancer treatment, the various nanoformulations, and actively describes heterocyclic magnetic nano catalysts and heterocyclic moiety, as well as their mode of action, which have favorable anti - cancer effects.
Collapse
Affiliation(s)
- Sankha Bhattacharya
- Department of Pharmaceutics, School of Pharmacy & Technology Management, SVKM\'S NMIMS Deemed-to-be University, Shirpur, Maharashtra 425405, India
| | - Rajat Patel
- Department of Pharmaceutics, School of Pharmacy & Technology Management, SVKM\'S NMIMS Deemed-to-be University, Shirpur, Maharashtra 425405, India
| | - Aalind Joshi
- Department of Pharmaceutics, School of Pharmacy & Technology Management, SVKM\'S NMIMS Deemed-to-be University, Shirpur, Maharashtra 425405, India
| |
Collapse
|
49
|
Liu Q. The emerging roles of exosomal long non-coding RNAs in bladder cancer. J Cell Mol Med 2022; 26:966-976. [PMID: 34981655 PMCID: PMC8831985 DOI: 10.1111/jcmm.17152] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 11/30/2021] [Accepted: 12/14/2021] [Indexed: 12/12/2022] Open
Abstract
Extracellular vesicles (EVs), especially exosomes, have been reported to play essential roles as extracellular messengers by transporting goods in various diseases, while their potential roles in bladder cancer (BC) still remain to be further studied. BC exhibits a high degree of chemoresistance and metastatic ability, which may be affected by cancer‐derived exosomes that carry proteins, lipids and RNA. To date, the most studied exosomal molecular cargo is long non‐coding RNA (lncRNA). Although there is increasing interest in its role and function, there is relatively little knowledge about it compared with other RNA transcripts. Nevertheless, in the past ten years, we have witnessed increasing interest in the role and function of lncRNA. For example, lncRNAs have been studied as potential biomarkers for the diagnosis of BC. They may play a role as a therapeutic target in precision medicine, but they may also be directly involved in the characteristics of tumour progression, such as metastasis, epithelial‐mesenchymal transition and drug resistance. Cancer cells are on chemotherapy acting. The function of lncRNA in various cancer exosomes has not yet been determined. In this review, we summarize the current studies about the prominent roles of exosomal lncRNAs in genome integrity, BC progression and carcinogenic features.
Collapse
Affiliation(s)
- Qiang Liu
- Department of Urology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning, China
| |
Collapse
|
50
|
Croft PKD, Sharma S, Godbole N, Rice GE, Salomon C. Ovarian-Cancer-Associated Extracellular Vesicles: Microenvironmental Regulation and Potential Clinical Applications. Cells 2021; 10:cells10092272. [PMID: 34571921 PMCID: PMC8471580 DOI: 10.3390/cells10092272] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 08/08/2021] [Accepted: 08/25/2021] [Indexed: 12/12/2022] Open
Abstract
Ovarian cancer (OC) is one of the most diagnosed gynecological cancers in women. Due to the lack of effective early stage screening, women are more often diagnosed at an advanced stage; therefore, it is associated with poor patient outcomes. There are a lack of tools to identify patients at the highest risk of developing this cancer. Moreover, early detection strategies, therapeutic approaches, and real-time monitoring of responses to treatment to improve survival and quality of life are also inadequate. Tumor development and progression are dependent upon cell-to-cell communication, allowing cancer cells to re-program cells not only within the surrounding tumor microenvironment, but also at distant sites. Recent studies established that extracellular vesicles (EVs) mediate bi-directional communication between normal and cancerous cells. EVs are highly stable membrane vesicles that are released from a wide range of cells, including healthy and cancer cells. They contain tissue-specific signaling molecules (e.g., proteins and miRNA) and, once released, regulate target cell phenotypes, inducing a pro-tumorigenic and immunosuppressive phenotype to contribute to tumor growth and metastasis as well as proximal and distal cell function. Thus, EVs are a “fingerprint” of their cell of origin and reflect the metabolic status. Additionally, via the capacity to evade the immune system and remain stable over long periods in circulation, EVs can be potent therapeutic agents. This review examines the potential role of EVs in the different aspects of the tumor microenvironment in OC, as well as their application in diagnosis, delivery of therapeutic agents, and disease monitoring.
Collapse
Affiliation(s)
- Priyakshi Kalita-de Croft
- Exosome Biology Laboratory, Centre for Clinical Diagnostics, University of Queensland Centre for Clinical Research, Royal Brisbane and Women’s Hospital, The University of Queensland, Herston, QLD 4029, Australia; (P.K.-d.C); (S.S); (N.G); (G.E.R)
- Faculty of Medicine, University of Queensland Centre for Clinical Research, Royal Brisbane and Women’s Hospital, The University of Queensland, Herston, QLD 4029, Australia
| | - Shayna Sharma
- Exosome Biology Laboratory, Centre for Clinical Diagnostics, University of Queensland Centre for Clinical Research, Royal Brisbane and Women’s Hospital, The University of Queensland, Herston, QLD 4029, Australia; (P.K.-d.C); (S.S); (N.G); (G.E.R)
| | - Nihar Godbole
- Exosome Biology Laboratory, Centre for Clinical Diagnostics, University of Queensland Centre for Clinical Research, Royal Brisbane and Women’s Hospital, The University of Queensland, Herston, QLD 4029, Australia; (P.K.-d.C); (S.S); (N.G); (G.E.R)
| | - Gregory E. Rice
- Exosome Biology Laboratory, Centre for Clinical Diagnostics, University of Queensland Centre for Clinical Research, Royal Brisbane and Women’s Hospital, The University of Queensland, Herston, QLD 4029, Australia; (P.K.-d.C); (S.S); (N.G); (G.E.R)
| | - Carlos Salomon
- Exosome Biology Laboratory, Centre for Clinical Diagnostics, University of Queensland Centre for Clinical Research, Royal Brisbane and Women’s Hospital, The University of Queensland, Herston, QLD 4029, Australia; (P.K.-d.C); (S.S); (N.G); (G.E.R)
- Correspondence: ; Tel.: +61-7-3346-5500; Fax: +61-7-3346-5509
| |
Collapse
|