1
|
Pal P, Sharma M, Gupta SK, Potdar MB, Belgamwar AV. miRNA-124 loaded extracellular vesicles encapsulated within hydrogel matrices for combating chemotherapy-induced neurodegeneration. Biochem Biophys Res Commun 2024; 734:150778. [PMID: 39368371 DOI: 10.1016/j.bbrc.2024.150778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Revised: 09/17/2024] [Accepted: 09/30/2024] [Indexed: 10/07/2024]
Abstract
Chemotherapy-induced neurodegeneration represents a significant challenge in cancer survivorship, manifesting in cognitive impairments that severely affect patients' quality of life. Emerging neuroregenerative therapies offer promise in mitigating these adverse effects, with miRNA-124 playing a pivotal role due to its critical functions in neural differentiation, neurogenesis, and neuroprotection. This review article delves into the innovative approach of using miRNA-124-loaded extracellular vesicles (EVs) encapsulated within hydrogel matrices as a targeted strategy for combating chemotherapy-induced neurodegeneration. We explore the biological underpinnings of miR-124 in neuroregeneration, detailing its mechanisms of action and therapeutic potential. The article further examines the roles and advantages of EVs as natural delivery systems for miRNAs and the application of hydrogel matrices in creating a sustained release environment conducive to neural tissue regeneration. By integrating these advanced materials and biological agents, we highlight a synergistic therapeutic strategy that leverages the bioactive properties of miR-124, the targeting capabilities of EVs, and the supportive framework of hydrogels. Preclinical studies and potential pathways to clinical translation are discussed, alongside the challenges, ethical considerations, and future directions in the field. This comprehensive review underscores the transformative potential of miR-124-loaded EVs in hydrogel matrices, offering insights into their development as a novel and integrative approach for addressing the complexities of chemotherapy-induced neurodegeneration.
Collapse
Affiliation(s)
- Pankaj Pal
- IIMT College of Pharmacy, IIMT Group of Colleges, Greater Noida, Uttar Pradesh, India.
| | - Monika Sharma
- Department of Pharmacy, Banasthali Vidyapith, Rajasthan, India
| | - Sukesh Kumar Gupta
- Department of Ophthalmology, Visual and Anatomical Sciences (OVAS), School of Medicine, Wayne State University, USA; KIET School of Pharmacy, KIET Group of Institutions, Ghaziabad, Uttar Pradesh, India
| | - Mrugendra B Potdar
- Department of Pharmaceutics, Shri Vile Parle Kelvani Mandal's Institute of Pharmacy, Dhule, Maharashtra, India
| | - Aarti V Belgamwar
- Department of Pharmaceutics, Shri Vile Parle Kelvani Mandal's Institute of Pharmacy, Dhule, Maharashtra, India
| |
Collapse
|
2
|
Gu X, He L, Zhang J, Xu H, Shen H, Huang R, Li Z. Recent Advances in Wash-Free Detection Methods of Extracellular Vesicles: A Mini Review. ACS Sens 2024. [PMID: 39446112 DOI: 10.1021/acssensors.4c00315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2024]
Abstract
Extracellular vesicles (EVs) are emerging biomarkers in liquid biopsy that have gained increasing attention in disease diagnosis and prognosis monitoring. Most reported detection methods require the isolation of EVs from complex body liquids, often involving multiple washing steps to remove excess reagents and eliminate background interference. Nonetheless, these methods not only cause the loss of EVs but also result in poor repeatability and prolonged detection duration. The focus on wash-free detection methods is increasing due to the specific ability to avoid the removal of surplus reagents and, in some cases, even the isolation and purification of EVs. Viewing from different methodological perspectives, this review summarizes the recent advances in wash-free detection of EVs, containing aggregation induction, proximity sensing, allosteric probes, phase separation, Roman spectroscopy, field-effect transistor and microcantilever. The pros and cons of each detection strategy are impartially evaluated and this review concludes the prospects for future developments in this field.
Collapse
Affiliation(s)
- Xinrui Gu
- Clinical Laboratory, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Zhongshan Road 321, Nanjing, Jiangsu Province 210008, China
| | - Lei He
- Clinical Laboratory, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Zhongshan Road 321, Nanjing, Jiangsu Province 210008, China
| | - Jinsong Zhang
- Clinical Laboratory, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Zhongshan Road 321, Nanjing, Jiangsu Province 210008, China
| | - Hongpan Xu
- Clinical Laboratory, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Zhongshan Road 321, Nanjing, Jiangsu Province 210008, China
| | - Han Shen
- Clinical Laboratory, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Zhongshan Road 321, Nanjing, Jiangsu Province 210008, China
| | - Rongrong Huang
- School of Pharmaceutical Sciences, Nanjing Tech University, South Puzhu Road 30, Nanjing, Jiangsu Province 211816, China
| | - Zhiyang Li
- Clinical Laboratory, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Zhongshan Road 321, Nanjing, Jiangsu Province 210008, China
| |
Collapse
|
3
|
Ibrahim UH, Gafar MA, Khan R, Tageldin A, Govender T, Mackraj I. Engineered extracellular vesicles coated with an antimicrobial peptide for advanced control of bacterial sepsis. JOURNAL OF EXTRACELLULAR BIOLOGY 2024; 3:e70000. [PMID: 39185334 PMCID: PMC11342353 DOI: 10.1002/jex2.70000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 07/15/2024] [Accepted: 08/01/2024] [Indexed: 08/27/2024]
Abstract
Alarming sepsis-related mortality rates present significant challenges to healthcare services globally. Despite advances made in the field, there is still an urgent need to develop innovative approaches that could improve survival rates and reduce the overall cost of treatment for sepsis patients. Therefore, this study aimed to develop a novel multifunctional therapeutic agent for advanced control of bacterial sepsis. Extracellular vesicles (EVs) isolated from lipopolysaccharide (LPS) induced HepG2 (hepatocellular carcinoma cells) (iEV) displayed an average particle size of 171.63 ± 2.77 nm, a poly dispersion index (PDI) of 0.32 ± 0.0, and a zeta potential (ZP) of -11.87 ± 0.18 mV. Compared to HepG2 EV, LPS induction significantly increases the EV protein concentration, PDI and ZP, reduces the average size and promotes cell proliferation and cytoprotective effects of the isolated EVs (iEVs) against LPS-induced cytotoxicity. Coating of iEV with a cationic antimicrobial peptide (AMP) to form PC-iEV slightly changed their physical properties and shifted their surface charge toward neutral values. This modification improved the antibacterial activity (2-fold lower minimum bactericidal concentration [MBC] values) and biocompatibility of the conjugated peptide while maintaining iEV cytoprotective and anti-inflammatory activities. Our findings indicate the superior anti-inflammatory and antibacterial dual activity of PC-iEV against pathogens associated with sepsis.
Collapse
Affiliation(s)
- Usri H. Ibrahim
- Discipline of Human Physiology, School of Laboratory Medicine and Medical Sciences, College of Health SciencesUniversity of KwaZulu‐NatalDurbanSouth Africa
| | - Mohammed A. Gafar
- Discipline of Pharmaceutical Sciences, College of Health SciencesUniversity of KwaZulu‐NatalDurbanSouth Africa
| | - Rene Khan
- Discipline of Medical Biochemistry, School of Laboratory Medicine and Medical ScienceUniversity of KwaZulu‐NatalDurbanSouth Africa
| | - Abdelrahman Tageldin
- Discipline of Pharmaceutical Sciences, College of Health SciencesUniversity of KwaZulu‐NatalDurbanSouth Africa
| | - Thirumala Govender
- Discipline of Pharmaceutical Sciences, College of Health SciencesUniversity of KwaZulu‐NatalDurbanSouth Africa
| | - Irene Mackraj
- Discipline of Human Physiology, School of Laboratory Medicine and Medical Sciences, College of Health SciencesUniversity of KwaZulu‐NatalDurbanSouth Africa
| |
Collapse
|
4
|
Wang C, Yan B, Liao P, Chen F, Lei P. Meta-Analysis of the Therapeutic Effects of Stem Cell-Derived Extracellular Vesicles in Rodent Models of Hemorrhagic Stroke. Stem Cells Int 2024; 2024:3390446. [PMID: 39263375 PMCID: PMC11390234 DOI: 10.1155/2024/3390446] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 03/28/2024] [Accepted: 04/17/2024] [Indexed: 09/13/2024] Open
Abstract
Background Stem cell-derived extracellular vesicles (SCEVs) have emerged as a potential therapy for hemorrhagic stroke. However, their effects are not fully understood. The aim of this study was to comprehensively evaluate the effects of SCEVs therapy in rodent models of hemorrhagic stroke, including subarachnoid hemorrhage (SAH) and intracerebral hemorrhage (ICH). Materials and Methods We conducted a comprehensive search of PubMed, EMBASE, and Web of Science until May 2023 to identify studies investigating the effects of SCEVs therapy in rodent models of ICH. The functional outcomes were assessed using neurobehavioral scores. Standardized mean differences (SMDs) and confidence intervals (CIs) were calculated using a random-effects model. Three authors independently screened the articles based on inclusion and exclusion criteria. All statistical analyses were performed using Revman 5.3 and Stata 17.0. Results Twelve studies published between 2018 and 2023 met the inclusion criteria. Our results showed that SCEVs therapy improved neurobehavioral scores in the rodent SAH model (SMD = -3.49, 95% CI: -4.23 to -2.75; p < 0.001). Additionally, SCEVs therapy improved the chronic neurobehavioral scores of the rodent ICH model (SMD = 2.38, 95% CI: 0.36-4.40; p=0.02) but did not have a significant impact on neurobehavioral scores in the acute and subacute phases. Significant heterogeneity was observed among the studies, and further stratification and sensitivity analyses failed to identify the source of heterogeneity. Conclusions Our findings suggest that SCEVs therapy may improve neurofunctional behavior after hemorrhagic stroke and provide important insights into the design of preclinical trials.
Collapse
Affiliation(s)
- Conglin Wang
- Department of Geriatrics Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Bo Yan
- Department of Geriatrics Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Pan Liao
- School of Medicine Nankai University, Tianjin 300071, China
| | - Fanglian Chen
- Department of Neurology Xuanwu Hospital Capital Medical University, Beijing 100053, China
| | - Ping Lei
- Department of Geriatrics Tianjin Medical University General Hospital, Tianjin 300052, China
| |
Collapse
|
5
|
Ding S, Kim YJ, Huang KY, Um D, Jung Y, Kong H. Delivery-mediated exosomal therapeutics in ischemia-reperfusion injury: advances, mechanisms, and future directions. NANO CONVERGENCE 2024; 11:18. [PMID: 38689075 PMCID: PMC11061094 DOI: 10.1186/s40580-024-00423-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Accepted: 04/05/2024] [Indexed: 05/02/2024]
Abstract
Ischemia-reperfusion injury (IRI) poses significant challenges across various organ systems, including the heart, brain, and kidneys. Exosomes have shown great potentials and applications in mitigating IRI-induced cell and tissue damage through modulating inflammatory responses, enhancing angiogenesis, and promoting tissue repair. Despite these advances, a more systematic understanding of exosomes from different sources and their biotransport is critical for optimizing therapeutic efficacy and accelerating the clinical adoption of exosomes for IRI therapies. Therefore, this review article overviews the administration routes of exosomes from different sources, such as mesenchymal stem cells and other somatic cells, in the context of IRI treatment. Furthermore, this article covers how the delivered exosomes modulate molecular pathways of recipient cells, aiding in the prevention of cell death and the promotions of regeneration in IRI models. In the end, this article discusses the ongoing research efforts and propose future research directions of exosome-based therapies.
Collapse
Affiliation(s)
- Shengzhe Ding
- Chemical & Biomolecular Engineering, University of Illinois, Urbana, IL, 61801, USA
| | - Yu-Jin Kim
- Center for Biomaterials, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
| | - Kai-Yu Huang
- Chemical & Biomolecular Engineering, University of Illinois, Urbana, IL, 61801, USA
| | - Daniel Um
- Bioengineering, University of Illinois, Urbana, IL, 61801, USA
| | - Youngmee Jung
- Center for Biomaterials, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
- Department of Electrical and Electronic Engineering, YU-KIST Institute, Yonsei University, Seoul, 03722, Republic of Korea
| | - Hyunjoon Kong
- Chemical & Biomolecular Engineering, University of Illinois, Urbana, IL, 61801, USA.
- Bioengineering, University of Illinois, Urbana, IL, 61801, USA.
- Carl R. Woese Institute for Genomic Biology, University of Illinois, Urbana, IL, 61801, USA.
- Chan Zuckerberg Biohub-Chicago, Chicago, USA.
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, Republic of Korea.
| |
Collapse
|
6
|
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
|
7
|
Singh G, Mehra A, Arora S, Gugulothu D, Vora LK, Prasad R, Khatri DK. Exosome-mediated delivery and regulation in neurological disease progression. Int J Biol Macromol 2024; 264:130728. [PMID: 38467209 DOI: 10.1016/j.ijbiomac.2024.130728] [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: 12/08/2023] [Revised: 03/05/2024] [Accepted: 03/06/2024] [Indexed: 03/13/2024]
Abstract
Exosomes (EXOs), membranous structures originating from diverse biological sources, have recently seized the attention of researchers due to their theranostic potential for neurological diseases. Released actively by various cells, including stem cells, adipose tissue, and immune cells, EXOs wield substantial regulatory influence over the intricate landscape of neurological complications, exhibiting both positive and negative modulatory effects. In AD, EXOs play a pivotal role in disseminating and breaking down amyloid-β protein. Moreover, EXOs derived from mesenchymal stem cells showcase a remarkable capacity to mitigate pro-inflammatory phenotypes by regulating miRNAs in neurodegenerative diseases. These vesicles possess the unique ability to traverse the blood-brain barrier, governing the aggregation of mutant huntingtin protein. Understanding the exosomal functions within the CNS holds significant promise for enhancing treatment efficacy in neurological diseases. This review intricately examines the regulatory mechanisms involving EXOs in neurological disease development, highlighting therapeutic prospects and exploring their utility in exosome-based nanomedicine for various neurological complications. Additionally, the review highlights the challenges associated with drug delivery to the brain, emphasizing the complexities inherent in this critical aspect of neurotherapeutics.
Collapse
Affiliation(s)
- Gurpreet Singh
- Molecular and cellular neuroscience lab, Department of pharmacology and toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Hyderabad, India
| | - Ankit Mehra
- Molecular and cellular neuroscience lab, Department of pharmacology and toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Hyderabad, India
| | - Sanchit Arora
- Department of Pharmaceutics, Delhi Institute of Pharmaceutical Sciences and Research (DIPSAR), Delhi Pharmaceutical Sciences and Research University (DPSRU), M.B. Road, Pushp Vihar, Sector-3, New Delhi 110017, India
| | - Dalapathi Gugulothu
- Department of Pharmaceutics, Delhi Institute of Pharmaceutical Sciences and Research (DIPSAR), Delhi Pharmaceutical Sciences and Research University (DPSRU), M.B. Road, Pushp Vihar, Sector-3, New Delhi 110017, India.
| | - Lalitkumar K Vora
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, BT9 7BL, UK.
| | - Renuka Prasad
- Department of Anatomy, Korea University College of Medicine, Moonsuk Medical Research Building, 516, 5th floor, 73 Inchon-ro, Seongbuk-gu, Seoul 02841, Republic of Korea
| | - Dharmendra Kumar Khatri
- Molecular and cellular neuroscience lab, Department of pharmacology and toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Hyderabad, India; Department of Pharmacology, Shobhaben Pratapbai Patel School of Pharmacy & Technology Management, SVKM's Narsee Monjee Institute of Management Studies (NMIMS) Deemed-to-University, Mumbai 400056, India.
| |
Collapse
|
8
|
Jiang J, Zhang X, Wang H, Spanos M, Jiang F, Ni L, Li J, Li G, Lin Y, Xiao J. Closer to The Heart: Harnessing the Power of Targeted Extracellular Vesicle Therapies. Adv Biol (Weinh) 2024; 8:e2300141. [PMID: 37953665 DOI: 10.1002/adbi.202300141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Revised: 09/08/2023] [Indexed: 11/14/2023]
Abstract
Extracellular vesicles (EVs) have emerged as novel diagnostic and therapeutic approaches for cardiovascular diseases. EVs derived from various origins exhibit distinct effects on the cardiovascular system. However, the application of native EVs is constrained due to their poor stabilities and limited targeting capabilities. Currently, targeted modification of EVs primarily involves genetic engineering, chemical modification (covalent, non-covalent), cell membrane modification, and biomaterial encapsulation. These techniques enhance the stability, biological activity, target-binding capacity, and controlled release of EVs at specific cells and tissues. The diverse origins of cardioprotective EVs are covered, and the applications of cardiac-targeting EV delivery systems in protecting against cardiovascular diseases are discussed. This review summarizes the current stage of research on the potential of EV-based targeted therapies for addressing cardiovascular disorders.
Collapse
Affiliation(s)
- Jizong Jiang
- Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Medicine, Shanghai University, Shanghai, 200444, China
| | - Xinxin Zhang
- Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Medicine, Shanghai University, Shanghai, 200444, China
| | - Hongyun Wang
- Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Medicine, Shanghai University, Shanghai, 200444, China
| | - Michail Spanos
- Cardiovascular Division of the Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
| | - Fei Jiang
- Department of Nursing, Union Hospital, Fujian Medical University Union Hospital, Fuzhou, 350001, China
- Department of Cardiovascular Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, China
| | - Lingyan Ni
- Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Medicine, Shanghai University, Shanghai, 200444, China
| | - Jin Li
- Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Medicine, Shanghai University, Shanghai, 200444, China
| | - Guoping Li
- Cardiovascular Division of the Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
| | - Yanjuan Lin
- Department of Nursing, Union Hospital, Fujian Medical University Union Hospital, Fuzhou, 350001, China
- Department of Cardiovascular Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, China
| | - Junjie Xiao
- Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Medicine, Shanghai University, Shanghai, 200444, China
| |
Collapse
|
9
|
Hao S, Yang H, Hu J, Luo L, Yuan Y, Liu L. Bioactive compounds and biological functions of medicinal plant-derived extracellular vesicles. Pharmacol Res 2024; 200:107062. [PMID: 38211637 DOI: 10.1016/j.phrs.2024.107062] [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: 09/08/2023] [Revised: 12/07/2023] [Accepted: 01/05/2024] [Indexed: 01/13/2024]
Abstract
Extracellular vesicles (EVs) are tiny lipid bilayer-enclosed membrane particles released from a variety of cell types into the surrounding environment. These EVs have massive participated in cell-to-cell communication and interspecies communication. In recent years, plant-derived extracellular vesicles (PDEVs) and "exosome-like" EVs populations found in distinct plants have attracted widespread attention. Especially, research on medicinal plant-derived extracellular vesicles (MPDEVs) are increasing, which are considered a kind of promising natural compound. This review summarizes current knowledge on MPDEVs in terms of bioactive compounds, including small RNA, protein, lipid, and metabolite, have been found on the surface and/or in the lumen of MPDEVs. Moreover, both in vitro and in vivo experiments have shown that MPDEVs exert broad biomedical functions, such as anti-inflammatory, anticancer, antioxidant, modulate microbiota, etc. MPDEVs may be a better substitute than animal-derived extracellular vesicles (ADEVs) because of safety and biocompatibility in the future.
Collapse
Affiliation(s)
- Siyu Hao
- National Resource Center for Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Hongyu Yang
- National Resource Center for Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China; Guangxi University of Chinese Medicine, School of Pharmacy, Nanning, China
| | - Jiaojiao Hu
- China Agricultural University, Department of Nutrition and Health, Beijing, China; Guangxi University of Chinese Medicine, School of Pharmacy, Nanning, China
| | - Lili Luo
- China Agricultural University, Department of Nutrition and Health, Beijing, China
| | - Yuan Yuan
- National Resource Center for Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China.
| | - Libing Liu
- China Agricultural University, Department of Nutrition and Health, Beijing, China.
| |
Collapse
|
10
|
Jing Z, Zhang G, Cai Y, Liang J, Lv L, Dang X. Engineered extracellular vesicle-delivered TGF-β inhibitor for attenuating osteoarthritis by targeting subchondral bone. J Tissue Eng 2024; 15:20417314241257781. [PMID: 39071897 PMCID: PMC11273819 DOI: 10.1177/20417314241257781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 05/13/2024] [Indexed: 07/30/2024] Open
Abstract
Osteoarthritis (OA) is a disease that affects the entire joint. To treat OA, it may be beneficial to inhibit the activity of TGF-β in the subchondral bone. However, delivering drugs to the subchondral bone using conventional methods is challenging. In this study, we developed an extracellular vesicle delivery system. The utilization of macrophage-derived extracellular vesicles as a drug-carrying platform enables drugs to evade immune clearance and cross biological barriers. By incorporating targeting peptides on the surface of extracellular vesicles, the drug platform becomes targeted. The combination of these two factors results in the successful delivery of the drug to the subchondral bone. The study evaluated the stability, cytotoxicity, and bone targeting capability of the engineered extracellular vesicle platform (BT-EV-G). It also assessed the effects of BT-EV-G on the differentiation, proliferation, and migration of bone mesenchymal stem cells (BMSCs). Additionally, the researchers administered BT-EV-G to anterior cruciate ligament transection (ACLT)-induced OA mice. The results showed that BT-EV-G had low toxicity and high bone targeting ability both in vitro and in vivo. BT-EV-G can restore coupled bone remodeling in subchondral bone by inhibiting pSmad2/3-dependent TGF-β signaling. This work provides new insights into the treatment of OA.
Collapse
Affiliation(s)
- Zhaopu Jing
- The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Guangyang Zhang
- The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Yuanqing Cai
- The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Jialin Liang
- The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Leifeng Lv
- The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Xiaoqian Dang
- The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| |
Collapse
|
11
|
Rosso G, Cauda V. Biomimicking Extracellular Vesicles with Fully Artificial Ones: A Rational Design of EV-BIOMIMETICS toward Effective Theranostic Tools in Nanomedicine. ACS Biomater Sci Eng 2023; 9:5924-5932. [PMID: 36535896 PMCID: PMC10646844 DOI: 10.1021/acsbiomaterials.2c01025] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 12/02/2022] [Indexed: 12/24/2022]
Abstract
Extracellular Vesicles (EVs) are the protagonists in cell communication and membrane trafficking, being responsible for the delivery of innumerable biomolecules and signaling moieties. At the moment, they are of paramount interest to researchers, as they naturally show incredibly high efficiency and specificity in delivering their cargo. For these reasons, EVs are employed or inspire the development of nanosized therapeutic delivery systems. In this Perspective, we propose an innovative strategy for the rational design of EV-mimicking vesicles (EV-biomimetics) for theranostic scopes. We first report on the current state-of-the-art use of EVs and their byproducts, such as surface-engineered EVs and EV-hybrids, having an artificial cargo (drug molecule, genetic content, nanoparticles, or dye incorporated in their lumen). Thereafter, we report on the new emerging field of EV-mimicking vesicles for theranostic scopes. We introduce an approach to prepare new, fully artificial EV-biomimetics, with particular attention to maintaining the natural reference lipidic composition. We overview those studies investigating natural EV membranes and the possible strategies to identify key proteins involved in site-selective natural homing, typical of EVs, and their cargo transfer to recipient cells. We propose the use also of molecular simulations, in particular of machine learning models, to approach the problem of lipid organization and self-assembly in natural EVs. We also discuss the beneficial feedback that could emerge combining the experimental tests with atomistic and molecular simulations when designing an EV-biomimetics lipid bilayer. The expectations from both research and industrial fields on fully artificial EV-biomimetics, having the same key functions of natural ones plus new diagnostic or therapeutic functions, could be enormous, as they can greatly expand the nanomedicine applications and guarantee on-demand and scalable production, off-the-shelf storage, high reproducibility of morphological and functional properties, and compliance with regulatory standards.
Collapse
Affiliation(s)
- Giada Rosso
- Department of Applied Science
and Technology, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Turin, Italy
| | - Valentina Cauda
- Department of Applied Science
and Technology, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Turin, Italy
| |
Collapse
|
12
|
Namini MS, Daneshimehr F, Beheshtizadeh N, Mansouri V, Ai J, Jahromi HK, Ebrahimi-Barough S. Cell-free therapy based on extracellular vesicles: a promising therapeutic strategy for peripheral nerve injury. Stem Cell Res Ther 2023; 14:254. [PMID: 37726794 PMCID: PMC10510237 DOI: 10.1186/s13287-023-03467-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 08/22/2023] [Indexed: 09/21/2023] Open
Abstract
Peripheral nerve injury (PNI) is one of the public health concerns that can result in a loss of sensory or motor function in the areas in which injured and non-injured nerves come together. Up until now, there has been no optimized therapy for complete nerve regeneration after PNI. Exosome-based therapies are an emerging and effective therapeutic strategy for promoting nerve regeneration and functional recovery. Exosomes, as natural extracellular vesicles, contain bioactive molecules for intracellular communications and nervous tissue function, which could overcome the challenges of cell-based therapies. Furthermore, the bioactivity and ability of exosomes to deliver various types of agents, such as proteins and microRNA, have made exosomes a potential approach for neurotherapeutics. However, the type of cell origin, dosage, and targeted delivery of exosomes still pose challenges for the clinical translation of exosome therapeutics. In this review, we have focused on Schwann cell and mesenchymal stem cell (MSC)-derived exosomes in nerve tissue regeneration. Also, we expressed the current understanding of MSC-derived exosomes related to nerve regeneration and provided insights for developing a cell-free MSC therapeutic strategy for nerve injury.
Collapse
Affiliation(s)
- Mojdeh Salehi Namini
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Regenerative Medicine Group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Fatemeh Daneshimehr
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Nima Beheshtizadeh
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Regenerative Medicine Group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Vahid Mansouri
- Digestive Disease Research Institute, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Jafar Ai
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Hossein Kargar Jahromi
- Research Center for Noncommunicable Diseases, Jahrom University of Medical Sciences, Jahrom, Iran.
| | - Somayeh Ebrahimi-Barough
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
13
|
Sakurai Y, Ohtani A, Nakayama Y, Gomi M, Masuda T, Ohtsuki S, Tanaka H, Akita H. Logistics and distribution of small extracellular vesicles from the subcutaneous space to the lymphatic system. J Control Release 2023; 361:77-86. [PMID: 37517544 DOI: 10.1016/j.jconrel.2023.07.043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 07/15/2023] [Accepted: 07/27/2023] [Indexed: 08/01/2023]
Abstract
Small extracellular vesicles (sEVs) are small, cell-derived particles with sizes of approximately 100 nm. Since these particles include cargos such as host cell-derived proteins, messenger RNAs, and micro RNAs, they serve as mediators of cell-cell communication. While the analysis of the pharmacokinetic of sEVs after the intravenous injection have been reported, the lymphatic transport of sEVs remains unclear. The objective of this study was to provide insights into the intra-lymphatic trafficking and distribution of sEVs when they are injected into an interstitial space both in normal skin tissue and in cancerous tissue. When sEVs were Subcutaneously administered into the tail base and the tumor tissue, they preferably accumulated in the lymph nodes (LNs), rather than in the liver and the spleen. The findings reported herein show that the lymphatic transport of sEVs was drastically changed in model mice, in which a surgical treatment was used to modify to allow the dominant lymphatic flow from the footpad directly to the axillary LN via the inguinal LN. Based on the results, we conclude that when sEVs are injected into the subcutis space, they are preferably delivered to the LN via the lymphatic system. Further, the extent of accumulation of sEVs in the LN after subcutaneous injection was reduced when they were preliminarily incubated with Proteinase K. These results suggest that the lymphatic drainage of sEVs in normal skin tissue is regulated by membrane proteins on their surface. This reduction, however, was not observed in the case of cancer tissue. This discrepancy can be attributed to the presence of highly permeable lymphatic vessels in the tumor tissue. Further, the major cell subtypes that captured sEVs in the LN were LN-resident medullary sinus macrophages. These collective findings indicate that the lymphatic drainage of sEVs are mediated by proteins and, that they may appear to contribute to the control of the function of immune-responsive cells in the LNs.
Collapse
Affiliation(s)
- Yu Sakurai
- Laboratory of DDS design and Drug Disposition, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan.
| | - Asa Ohtani
- Laboratory of DDS Design and Drug Disposition, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba 260-8675, Japan
| | - Yuka Nakayama
- Laboratory of DDS design and Drug Disposition, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan
| | - Masaki Gomi
- Laboratory of DDS Design and Drug Disposition, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba 260-8675, Japan
| | - Takeshi Masuda
- Laboratory of Pharmaceutical Microbiology, Faculty of Life Sciences, Kumamoto University, Kumamoto 862-0973, Japan
| | - Sumio Ohtsuki
- Laboratory of Pharmaceutical Microbiology, Faculty of Life Sciences, Kumamoto University, Kumamoto 862-0973, Japan
| | - Hiroki Tanaka
- Laboratory of DDS Design and Drug Disposition, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba 260-8675, Japan
| | - Hidetaka Akita
- Laboratory of DDS design and Drug Disposition, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan.
| |
Collapse
|
14
|
Danilushkina AA, Emene CC, Barlev NA, Gomzikova MO. Strategies for Engineering of Extracellular Vesicles. Int J Mol Sci 2023; 24:13247. [PMID: 37686050 PMCID: PMC10488046 DOI: 10.3390/ijms241713247] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 08/14/2023] [Accepted: 08/17/2023] [Indexed: 09/10/2023] Open
Abstract
Extracellular vesicles (EVs) are membrane vesicles released by cells into the extracellular space. EVs mediate cell-to-cell communication through local and systemic transportation of biomolecules such as DNA, RNA, transcription factors, cytokines, chemokines, enzymes, lipids, and organelles within the human body. EVs gained a particular interest from cancer biology scientists because of their role in the modulation of the tumor microenvironment through delivering bioactive molecules. In this respect, EVs represent an attractive therapeutic target and a means for drug delivery. The advantages of EVs include their biocompatibility, small size, and low immunogenicity. However, there are several limitations that restrict the widespread use of EVs in therapy, namely, their low specificity and payload capacity. Thus, in order to enhance the therapeutic efficacy and delivery specificity, the surface and composition of extracellular vesicles should be modified accordingly. In this review, we describe various approaches to engineering EVs, and further discuss their advantages and disadvantages to promote the application of EVs in clinical practice.
Collapse
Affiliation(s)
- Anna A. Danilushkina
- Laboratory of Intercellular Communications, Institute of Fundamental Medicine and Biology, Kazan Federal University, 420021 Kazan, Russia
| | - Charles C. Emene
- Laboratory of Intercellular Communications, Institute of Fundamental Medicine and Biology, Kazan Federal University, 420021 Kazan, Russia
| | - Nicolai A. Barlev
- Laboratory of Molecular Immunology, Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia
- Department of Biomedicine, Nazarbayev University School of Medicine, Astana 001000, Kazakhstan
| | - Marina O. Gomzikova
- Laboratory of Intercellular Communications, Institute of Fundamental Medicine and Biology, Kazan Federal University, 420021 Kazan, Russia
- Laboratory of Molecular Immunology, Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia
| |
Collapse
|
15
|
Zhang W, Ruan L. Recent advances in poor HIV immune reconstitution: what will the future look like? Front Microbiol 2023; 14:1236460. [PMID: 37608956 PMCID: PMC10440441 DOI: 10.3389/fmicb.2023.1236460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 07/24/2023] [Indexed: 08/24/2023] Open
Abstract
Combination antiretroviral therapy has demonstrated proved effectiveness in suppressing viral replication and significantly recovering CD4+ T cell count in HIV type-1 (HIV-1)-infected patients, contributing to a dramatic reduction in AIDS morbidity and mortality. However, the factors affecting immune reconstitution are extremely complex. Demographic factors, co-infection, baseline CD4 cell level, abnormal immune activation, and cytokine dysregulation may all affect immune reconstitution. According to report, 10-40% of HIV-1-infected patients fail to restore the normalization of CD4+ T cell count and function. They are referred to as immunological non-responders (INRs) who fail to achieve complete immune reconstitution and have a higher mortality rate and higher risk of developing other non-AIDS diseases compared with those who achieve complete immune reconstitution. Heretofore, the mechanisms underlying incomplete immune reconstitution in HIV remain elusive, and INRs are not effectively treated or mitigated. This review discusses the recent progress of mechanisms and factors responsible for incomplete immune reconstitution in AIDS and summarizes the corresponding therapeutic strategies according to different mechanisms to improve the individual therapy.
Collapse
Affiliation(s)
| | - Lianguo Ruan
- Department of Infectious Diseases, Wuhan Jinyintan Hospital, Tongji Medical College of Huazhong University of Science and Technology, Hubei Clinical Research Center for Infectious Diseases, Wuhan Research Center for Communicable Disease Diagnosis and Treatment, Chinese Academy of Medical Sciences, Joint Laboratory of Infectious Diseases and Health, Wuhan Institute of Virology and Wuhan Jinyintan Hospital, Chinese Academy of Sciences, Wuhan, Hubei, China
| |
Collapse
|
16
|
Das K, Paul S, Mukherjee T, Ghosh A, Sharma A, Shankar P, Gupta S, Keshava S, Parashar D. Beyond Macromolecules: Extracellular Vesicles as Regulators of Inflammatory Diseases. Cells 2023; 12:1963. [PMID: 37566042 PMCID: PMC10417494 DOI: 10.3390/cells12151963] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 07/26/2023] [Accepted: 07/27/2023] [Indexed: 08/12/2023] Open
Abstract
Inflammation is the defense mechanism of the immune system against harmful stimuli such as pathogens, toxic compounds, damaged cells, radiation, etc., and is characterized by tissue redness, swelling, heat generation, pain, and loss of tissue functions. Inflammation is essential in the recruitment of immune cells at the site of infection, which not only aids in the elimination of the cause, but also initiates the healing process. However, prolonged inflammation often brings about several chronic inflammatory disorders; hence, a balance between the pro- and anti-inflammatory responses is essential in order to eliminate the cause while producing the least damage to the host. A growing body of evidence indicates that extracellular vesicles (EVs) play a major role in cell-cell communication via the transfer of bioactive molecules in the form of proteins, lipids, DNA, RNAs, miRNAs, etc., between the cells. The present review provides a brief classification of the EVs followed by a detailed description of how EVs contribute to the pathogenesis of various inflammation-associated diseases and their implications as a therapeutic measure. The latter part of the review also highlights how EVs act as a bridging entity in blood coagulation disorders and associated inflammation. The findings illustrated in the present review may open a new therapeutic window to target EV-associated inflammatory responses, thereby minimizing the negative outcomes.
Collapse
Affiliation(s)
- Kaushik Das
- Department of Cellular and Molecular Biology, The University of Texas at Tyler Health Science Center, Tyler, TX 75708, USA
| | - Subhojit Paul
- School of Biological Sciences, Indian Association for the Cultivation of Science, Jadavpur, Kolkata 700032, India; (S.P.); (A.G.)
| | - Tanmoy Mukherjee
- School of Medicine, The University of Texas at Tyler Health Science Center, Tyler, TX 75708, USA;
| | - Arnab Ghosh
- School of Biological Sciences, Indian Association for the Cultivation of Science, Jadavpur, Kolkata 700032, India; (S.P.); (A.G.)
| | - Anshul Sharma
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01605, USA;
| | - Prem Shankar
- Department of Neurobiology, The University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555, USA;
| | - Saurabh Gupta
- Department of Biotechnology, GLA University, Mathura 281406, India;
| | - Shiva Keshava
- Department of Cellular and Molecular Biology, The University of Texas at Tyler Health Science Center, Tyler, TX 75708, USA
| | - Deepak Parashar
- Department of Medicine, Division of Hematology & Oncology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| |
Collapse
|
17
|
Wang J, Liu Y, Liu F, Gan S, Roy S, Hasan I, Zhang B, Guo B. Emerging extracellular vesicle-based carriers for glioblastoma diagnosis and therapy. NANOSCALE 2023. [PMID: 37337814 DOI: 10.1039/d3nr01667f] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2023]
Abstract
Glioblastoma (GBM) treatment is still a big clinical challenge because of its highly malignant, invasive, and lethal characteristics. After treatment with the conventional therapeutic paradigm of surgery combined with radio- and chemotherapy, patients bearing GBMs generally exhibit a poor prognosis, with high mortality and a high disability rate. The main reason is the existence of the formidable blood-brain barrier (BBB), aggressive growth, and the infiltration nature of GBMs. Especially, the BBB suppresses the delivery of imaging and therapeutic agents to lesion sites, and thus this leads to difficulties in achieving a timely diagnosis and treatment. Recent studies have demonstrated that extracellular vesicles (EVs) exhibit favorable merits including good biocompatibility, a strong drug loading capacity, long circulation time, good BBB crossing efficiency, specific targeting to lesion sites, and high efficiency in the delivery of a variety of cargos for GBM therapy. Importantly, EVs inherit physiological and pathological molecules from the source cells, which are ideal biomarkers for molecularly tracking the malignant progression of GBMs. Herein, we start by introducing the pathophysiology and physiology of GBMs, followed by presenting the biological functions of EVs in GBMs with a special focus on their role as biomarkers for GBM diagnosis and as messengers in the modulation of the GBM microenvironment. Furthermore, we provide an update on the recent progress of using EVs in biology, functionality, and isolation applications. More importantly, we systematically summarize the most recent advances of EV-based carriers for GBM therapy by delivering different drugs including gene/RNA-based drugs, chemotherapy drugs, imaging agents, and combinatory drugs. Lastly, we point out the challenges and prospects of future research on EVs for diagnosing and treating GBMs. We hope this review will stimulate interest from researchers with different backgrounds and expedite the progress of GBM treatment paradigms.
Collapse
Affiliation(s)
- Jingjing Wang
- Shenzhen Key Laboratory of Advanced Functional Carbon Materials Research and Comprehensive Application, Shenzhen Key Laboratory of Flexible Printed Electronics Technology, and School of Science, Harbin Institute of Technology, Shenzhen 518055, China.
- Shenzhen Key Laboratory of Flexible Printed Electronics Technology, Harbin Institute of Technology, Shenzhen 518055, China
| | - Yue Liu
- Shenzhen Key Laboratory of Advanced Functional Carbon Materials Research and Comprehensive Application, Shenzhen Key Laboratory of Flexible Printed Electronics Technology, and School of Science, Harbin Institute of Technology, Shenzhen 518055, China.
- Shenzhen Key Laboratory of Flexible Printed Electronics Technology, Harbin Institute of Technology, Shenzhen 518055, China
| | - Fengbo Liu
- Shenzhen Key Laboratory of Advanced Functional Carbon Materials Research and Comprehensive Application, Shenzhen Key Laboratory of Flexible Printed Electronics Technology, and School of Science, Harbin Institute of Technology, Shenzhen 518055, China.
- Shenzhen Key Laboratory of Flexible Printed Electronics Technology, Harbin Institute of Technology, Shenzhen 518055, China
| | - Shaoyan Gan
- Shenzhen Key Laboratory of Advanced Functional Carbon Materials Research and Comprehensive Application, Shenzhen Key Laboratory of Flexible Printed Electronics Technology, and School of Science, Harbin Institute of Technology, Shenzhen 518055, China.
- Shenzhen Key Laboratory of Flexible Printed Electronics Technology, Harbin Institute of Technology, Shenzhen 518055, China
| | - Shubham Roy
- Shenzhen Key Laboratory of Advanced Functional Carbon Materials Research and Comprehensive Application, Shenzhen Key Laboratory of Flexible Printed Electronics Technology, and School of Science, Harbin Institute of Technology, Shenzhen 518055, China.
- Shenzhen Key Laboratory of Flexible Printed Electronics Technology, Harbin Institute of Technology, Shenzhen 518055, China
| | - Ikram Hasan
- Shenzhen Key Laboratory of Advanced Functional Carbon Materials Research and Comprehensive Application, Shenzhen Key Laboratory of Flexible Printed Electronics Technology, and School of Science, Harbin Institute of Technology, Shenzhen 518055, China.
- Shenzhen Key Laboratory of Flexible Printed Electronics Technology, Harbin Institute of Technology, Shenzhen 518055, China
| | - Baozhu Zhang
- Department of Oncology, People's Hospital of Shenzhen Baoan District, The Second Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong 518055, China.
| | - Bing Guo
- Shenzhen Key Laboratory of Advanced Functional Carbon Materials Research and Comprehensive Application, Shenzhen Key Laboratory of Flexible Printed Electronics Technology, and School of Science, Harbin Institute of Technology, Shenzhen 518055, China.
- Shenzhen Key Laboratory of Flexible Printed Electronics Technology, Harbin Institute of Technology, Shenzhen 518055, China
| |
Collapse
|
18
|
Kronstadt SM, Van Heyningen LH, Aranda A, Jay SM. Assessment of anti-inflammatory bioactivity of extracellular vesicles is susceptible to error via media component contamination. Cytotherapy 2023; 25:387-396. [PMID: 36599771 PMCID: PMC10006399 DOI: 10.1016/j.jcyt.2022.12.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 12/01/2022] [Accepted: 12/12/2022] [Indexed: 01/03/2023]
Abstract
Extracellular vesicles (EVs) are widely implicated as novel diagnostic and therapeutic modalities for a wide range of diseases. Thus, optimization of EV biomanufacturing is of high interest. In the course of developing parameters for a human embryonic kidney cells (HEK293T) EV production platform, we examined the combinatorial effects of cell culture conditions (i.e., static versus dynamic) and isolation techniques (i.e., ultracentrifugation versus tangential flow filtration versus size-exclusion chromatography) on functional characteristics of HEK293T EVs, including anti-inflammatory bioactivity using a well-established lipopolysaccharide-stimulated mouse macrophage model. We unexpectedly found that, depending on culture condition and isolation strategy, HEK293T EVs appeared to significantly suppress the secretion of pro-inflammatory cytokines (i.e., interleukin-6, RANTES [regulated upon activation, normal T cell expressed and secreted]) in the stimulated mouse macrophages. Further examination revealed that these results were most likely due to non-EV fetal bovine serum components in HEK293T EV preparations. Thus, future research assessing the anti-inflammatory effects of EVs should be designed to account for this phenomenon.
Collapse
Affiliation(s)
- Stephanie M Kronstadt
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland, USA
| | | | - Amaya Aranda
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland, USA
| | - Steven M Jay
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland, USA; Program in Molecular and Cell Biology, University of Maryland, College Park, Maryland, USA.
| |
Collapse
|
19
|
Qi Y, Zhao X, Dong Y, Wang M, Wang J, Fan Z, Weng Q, Yu H, Li J. Opportunities and challenges of natural killer cell-derived extracellular vesicles. Front Bioeng Biotechnol 2023; 11:1122585. [PMID: 37064251 PMCID: PMC10102538 DOI: 10.3389/fbioe.2023.1122585] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 03/23/2023] [Indexed: 04/03/2023] Open
Abstract
Extracellular vesicles (EVs) are increasingly recognized as important intermediaries of intercellular communication. They have significant roles in many physiological and pathological processes and show great promise as novel biomarkers of disease, therapeutic agents, and drug delivery tools. Existing studies have shown that natural killer cell-derived EVs (NEVs) can directly kill tumor cells and participate in the crosstalk of immune cells in the tumor microenvironment. NEVs own identical cytotoxic proteins, cytotoxic receptors, and cytokines as NK cells, which is the biological basis for their application in antitumor therapy. The nanoscale size and natural targeting property of NEVs enable precisely killing tumor cells. Moreover, endowing NEVs with a variety of fascinating capabilities via common engineering strategies has become a crucial direction for future research. Thus, here we provide a brief overview of the characteristics and physiological functions of the various types of NEVs, focusing on their production, isolation, functional characterization, and engineering strategies for their promising application as a cell-free modality for tumor immunotherapy.
Collapse
Affiliation(s)
- Yuchen Qi
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Department of Oncology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Xiang Zhao
- Department of Oncology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
- *Correspondence: Xiang Zhao, ; Hua Yu, ; Jianjun Li,
| | - Yan Dong
- Department of Oncology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Min Wang
- Department of General Surgery, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Junyi Wang
- Department of General Surgery, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zhichao Fan
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Department of Oncology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Qin Weng
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Department of Oncology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Hua Yu
- Department of General Surgery, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
- *Correspondence: Xiang Zhao, ; Hua Yu, ; Jianjun Li,
| | - Jianjun Li
- Department of Oncology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
- *Correspondence: Xiang Zhao, ; Hua Yu, ; Jianjun Li,
| |
Collapse
|
20
|
Zhang W, Wang L, Guo H, Chen L, Huang X. Dapagliflozin-Loaded Exosome Mimetics Facilitate Diabetic Wound Healing by HIF-1α-Mediated Enhancement of Angiogenesis. Adv Healthc Mater 2023; 12:e2202751. [PMID: 36442997 PMCID: PMC11469070 DOI: 10.1002/adhm.202202751] [Citation(s) in RCA: 24] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Indexed: 11/30/2022]
Abstract
Angiogenesis plays a critical role in diabetic wound healing. However, no effective strategies have been developed to target endothelial cells (ECs) to facilitate diabetic wound healing. Dapagliflozin (DA) as a sodium-glucose linked transporter 2 (SGLT2) inhibitor, may promote neovascularization in diabetic mice via HIF-1α-mediated enhancement of angiogenesis. Here, the bioinspired nanovesicles (NVs) prepared from induced pluripotent stem cells-derived ECs through an extrusion approach are reported, which can function as exosome mimetics to achieve targeted deliver of DA. Abundant membrane C-X-C motif chemokine receptor 4 conferred the EC-targeting ability of these NVs and the endothelial homology facilitated the accumulation in ECs. Furthermore, these DA-loaded induced pluripotent stem cells (iPSC)-EC NVs can facilitate angiogenesis and diabetic wound healing by HIF-1α/VEGFA pathway. Taken together, this study indicated that targeting ECs and regulating angiogenesis may be a promising strategy for the treatment of diabetic wound healing.
Collapse
Affiliation(s)
- Weiyue Zhang
- Department of EndocrinologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
| | - Lutong Wang
- Department of OrthopaedicsUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
| | - Haoyu Guo
- Department of OrthopaedicsUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
| | - Lulu Chen
- Department of EndocrinologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
| | - Xin Huang
- Department of OrthopaedicsUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
| |
Collapse
|
21
|
Suthar J, Taub M, Carney RP, Williams GR, Guldin S. Recent developments in biosensing methods for extracellular vesicle protein characterization. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2023; 15:e1839. [PMID: 35999185 PMCID: PMC10078591 DOI: 10.1002/wnan.1839] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 06/27/2022] [Accepted: 07/13/2022] [Indexed: 01/31/2023]
Abstract
Research into extracellular vesicles (EVs) has grown significantly over the last few decades with EVs being widely regarded as a source of biomarkers for human health and disease with massive clinical potential. Secreted by every cell type in the body, EVs report on the internal cellular conditions across all tissue types. Their presence in readily accessible biofluids makes the potential of EV biosensing highly attractive as a noninvasive diagnostic platform via liquid biopsies. However, their small size (50-250 nm), inherent heterogeneity, and the complexity of the native biofluids introduce challenges for effective characterization, thus, limiting their clinical utility. This has led to a surge in the development of various novel EV biosensing techniques, with capabilities beyond those of conventional methods that have been directly transferred from cell biology. In this review, key detection principles used for EV biosensing are summarized, with a focus on some of the most recent and fundamental developments in the field over the last 5 years. This article is categorized under: Diagnostic Tools > Biosensing Diagnostic Tools > In Vitro Nanoparticle-Based Sensing.
Collapse
Affiliation(s)
- Jugal Suthar
- Department of Chemical Engineering, University College London, London, UK.,UCL School of Pharmacy, University College London, London, UK
| | - Marissa Taub
- UCL School of Pharmacy, University College London, London, UK
| | - Randy P Carney
- Department of Biomedical Engineering, University of California, Davis, Davis, California, USA
| | | | - Stefan Guldin
- Department of Chemical Engineering, University College London, London, UK
| |
Collapse
|
22
|
Das R, Kanjilal P, Medeiros J, Thayumanavan S. What's Next after Lipid Nanoparticles? A Perspective on Enablers of Nucleic Acid Therapeutics. Bioconjug Chem 2022; 33:1996-2007. [PMID: 35377622 PMCID: PMC9530067 DOI: 10.1021/acs.bioconjchem.2c00058] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Recent success of mRNA-based COVID-19 vaccines have bolstered the strength of nucleic acids as a therapeutic platform. The number of new clinical trial candidates is skyrocketing with the potential to address many unmet clinical needs. Despite advancements in other aspects, the systemic delivery of nucleic acids to target sites remains a major challenge. Thus, nucleic acid based therapy has yet to reach its full potential. In this review, we shed light on a select few prospective technologies that exhibit substantial potential over traditional nanocarrier designs for nucleic acid delivery. We critically analyze these systems with specific attention to the possibilities for clinical translation.
Collapse
Affiliation(s)
- Ritam Das
- Department of Chemistry, University of Massachusetts, Amherst, Massachusetts 01003, United States
- The Center for Bioactive Delivery-Institute for Applied Life Sciences, University of Massachusetts, Amherst, Massachusetts 01003, United States
| | - Pintu Kanjilal
- Department of Chemistry, University of Massachusetts, Amherst, Massachusetts 01003, United States
- The Center for Bioactive Delivery-Institute for Applied Life Sciences, University of Massachusetts, Amherst, Massachusetts 01003, United States
| | - Jewel Medeiros
- Department of Chemistry, University of Massachusetts, Amherst, Massachusetts 01003, United States
- The Center for Bioactive Delivery-Institute for Applied Life Sciences, University of Massachusetts, Amherst, Massachusetts 01003, United States
| | - S Thayumanavan
- Department of Chemistry, University of Massachusetts, Amherst, Massachusetts 01003, United States
- Department of Biomedical Engineering, University of Massachusetts, Amherst, Massachusetts 01003, United States
- Molecular and Cellular Biology Program, University of Massachusetts, Amherst, Massachusetts 01003, United States
- The Center for Bioactive Delivery-Institute for Applied Life Sciences, University of Massachusetts, Amherst, Massachusetts 01003, United States
| |
Collapse
|
23
|
Extracellular vesicle isolation, purification and evaluation in cancer diagnosis. Expert Rev Mol Med 2022; 24:e41. [PMID: 36268744 DOI: 10.1017/erm.2022.34] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Strategies for non-invasive biomarker discovery in early detection of cancer are an urgent need. Extracellular vesicles (EVs) have generated increasing attention from the scientific community and are under intensive investigations due to their unique biological profiles and their non-invasive nature. EVs are membrane-enclosed vesicles with variable sizes and function. Such vesicles are actively secreted from multiple cell types and are considered as key vehicles for inter-cellular communications and signalling. The stability and potential to easily cross biological barriers enable EVs for exerting durable effects on target cells. These along with easy access to such vesicles, the consistent secretion from tumour during all stages of tumorigenesis and their content providing a reservoir of molecules as well as mirroring the identity of the cell of origin are virtues that have made EVs appealing to be assessed in liquid biopsy approaches and for using as a promising resource of biomarkers in cancer diagnosis and therapy and monitoring targeted cancer therapy. Early detection of EVs will guide time-scheduled personalised therapy. Surveying reliable and sensitive methods for rapid isolation of EVs from biofluids, the purity of isolated vesicles and their molecular profiling and marker specification for clinical translation in patients with cancer are issues in the area and the hot topics of many recent studies. Here, the focus is over methods for EV isolation and stratification for digging more information about liquid biopsy-based diagnosis. Extending knowledge regarding EV-based strategies is a key to validate independent patient follow-up for cancer diagnosis at early stages and inspecting the efficacy of therapeutics.
Collapse
|
24
|
Engineered extracellular vesicles: Regulating the crosstalk between the skeleton and immune system. ENGINEERED REGENERATION 2022. [DOI: 10.1016/j.engreg.2022.06.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
|
25
|
Zhang K, Liu L, Shi K, Zhang K, Zheng C, Jin Y. Extracellular Vesicles for Immunomodulation in Tissue Regeneration. Tissue Eng Part C Methods 2022; 28:393-404. [PMID: 35856810 DOI: 10.1089/ten.tec.2022.0000059] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
A large number of people suffer from tissue injury and defect worldwide, which constitutes a critical challenge for regenerative medicine. During the complicated process of tissue repair and regeneration, immune response that involves many kinds of immune cells often concurrently exists and plays a significant role, thus providing a promising target for the development of therapeutic strategies. As a critical player in cell-cell communication, extracellular vesicles (EVs) are a cluster of nano-sized vesicles of different categories, which have been reported to possess favorable immunoregulatory potential, and participate in the process of tissue repair and regeneration. Furthermore, EVs can be engineered with genetic or chemical strategies for optimized performance as therapeutic mediators. Here, we provide an outline on the biology of EVs as well as the role of EVs in immune regulation, focusing on exosomes, microvesicles, and apoptotic vesicles. We further summarize the applications of EV-based therapies for tissue regeneration, with particular emphasis on the modulation of immune system. Also, we have discussed the construction strategies of engineered EVs and the immunomodulatory capability of engineered EVs as well as their therapeutic potential in tissue repair. This review will highlight the outstanding potential of EV-based therapeutic strategies for tissue repair and regeneration.
Collapse
Affiliation(s)
- Kaichao Zhang
- State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, China.,Xi'an Institute of Tissue Engineering and Regenerative Medicine, Xi'an, China
| | - Lu Liu
- State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, China.,Department of Orthodontics, School of Stomatology, The Fourth Military Medical University, Xi'an, China
| | - Ke Shi
- State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, China.,Xi'an Institute of Tissue Engineering and Regenerative Medicine, Xi'an, China
| | - Kai Zhang
- State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, China.,Department of Oral and Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, Xi'an, China
| | - Chenxi Zheng
- State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, China
| | - Yan Jin
- State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, China.,Xi'an Institute of Tissue Engineering and Regenerative Medicine, Xi'an, China
| |
Collapse
|
26
|
He N, Thippabhotla S, Zhong C, Greenberg Z, Xu L, Pessetto Z, Godwin AK, Zeng Y, He M. Nano pom-poms prepared exosomes enable highly specific cancer biomarker detection. Commun Biol 2022; 5:660. [PMID: 35787656 PMCID: PMC9253007 DOI: 10.1038/s42003-022-03598-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 06/16/2022] [Indexed: 01/27/2023] Open
Abstract
Extracellular vesicles (EVs), particularly nano-sized small EV exosomes, are emerging biomarker sources. However, due to heterogeneous populations secreted from diverse cell types, mapping exosome multi-omic molecular information specifically to their pathogenesis origin for cancer biomarker identification is still extraordinarily challenging. Herein, we introduced a novel 3D-structured nanographene immunomagnetic particles (NanoPoms) with unique flower pom-poms morphology and photo-click chemistry for specific marker-defined capture and release of intact exosome. This specific exosome isolation approach leads to the expanded identification of targetable cancer biomarkers with enhanced specificity and sensitivity, as demonstrated by multi-omic exosome analysis of bladder cancer patient tissue fluids using the next generation sequencing of somatic DNA mutations, miRNAs, and the global proteome (Data are available via ProteomeXchange with identifier PXD034454). The NanoPoms prepared exosomes also exhibit distinctive in vivo biodistribution patterns, highlighting the highly viable and integral quality. The developed method is simple and straightforward, which is applicable to nearly all types of biological fluids and amenable for enrichment, scale up, and high-throughput exosome isolation.
Collapse
Affiliation(s)
- Nan He
- Department of Chemical and Petroleum Engineering, Bioengineering Program, University of Kansas, Lawrence, KS, 66045, USA
- Clara Biotech Inc., Lawrence, KS, 66047, USA
| | - Sirisha Thippabhotla
- Department of Electrical Engineering and Computer Science, University of Kansas, Lawrence, KS, 66045, USA
| | - Cuncong Zhong
- Department of Electrical Engineering and Computer Science, University of Kansas, Lawrence, KS, 66045, USA
| | - Zachary Greenberg
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, FL, 32610, USA
| | - Liang Xu
- Department of Molecular Biosciences, University of Kansas, Lawrence, KS, 66045, USA
| | - Ziyan Pessetto
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Andrew K Godwin
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, 66160, USA
- University of Kansas Cancer Center, Kansas City, KS, 66160, USA
| | - Yong Zeng
- Department of Chemistry, University of Florida, Gainesville, FL, 32603, USA
| | - Mei He
- Department of Chemical and Petroleum Engineering, Bioengineering Program, University of Kansas, Lawrence, KS, 66045, USA.
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, FL, 32610, USA.
| |
Collapse
|
27
|
Tang TT, Wang B, Lv LL, Dong Z, Liu BC. Extracellular vesicles for renal therapeutics: State of the art and future perspective. J Control Release 2022; 349:32-50. [PMID: 35779658 DOI: 10.1016/j.jconrel.2022.06.049] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Revised: 06/24/2022] [Accepted: 06/25/2022] [Indexed: 12/21/2022]
Abstract
With the ever-increasing burden of kidney disease, the need for developing new therapeutics to manage this disease has never been greater. Extracellular vesicles (EVs) are natural membranous nanoparticles present in virtually all organisms. Given their excellent delivery capacity in the body, EVs have emerged as a frontier technology for drug delivery and have the potential to usher in a new era of nanomedicine for kidney disease. This review is focused on why EVs are such compelling drug carriers and how to release their fullest potentiality in renal therapeutics. We discuss the unique features of EVs compared to artificial nanoparticles and outline the engineering technologies and steps in developing EV-based therapeutics, with an emphasis on the emerging approaches to target renal cells and prolong kidney retention. We also explore the applications of EVs as natural therapeutics or as drug carriers in the treatment of renal disorders and present our views on the critical challenges in manufacturing EVs as next-generation renal therapeutics.
Collapse
Affiliation(s)
- Tao-Tao Tang
- Institute of Nephrology, Zhong Da Hospital, Nanjing, China; Department of Pathology and Pathophysiology, Southeast University School of Medicine, Nanjing, China
| | - Bin Wang
- Institute of Nephrology, Zhong Da Hospital, Nanjing, China
| | - Lin-Li Lv
- Institute of Nephrology, Zhong Da Hospital, Nanjing, China.
| | - Zheng Dong
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University and Charlie Norwood VA Medical Center, Augusta, GA, USA
| | - Bi-Cheng Liu
- Institute of Nephrology, Zhong Da Hospital, Nanjing, China.
| |
Collapse
|
28
|
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: 16.5] [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
|
29
|
Preparation, Characterization, and In Vitro Anticancer Activity Evaluation of Broccoli-Derived Extracellular Vesicle-Coated Astaxanthin Nanoparticles. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27123955. [PMID: 35745077 PMCID: PMC9230617 DOI: 10.3390/molecules27123955] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Revised: 06/01/2022] [Accepted: 06/09/2022] [Indexed: 11/16/2022]
Abstract
Astaxanthin (AST) is a type of ketone carotenoid having significant antioxidation and anticancer abilities. However, its application is limited due to its low stability and bioavailability. In our study, poly (lactic-co-glycolic acid) (PLGA)-encapsulated AST (AST@PLGA) nanoparticles were prepared by emulsion solvent evaporation method and then further processed by ultrasound with broccoli-derived extracellular vesicles (BEVs), thereby evolving as BEV-coated AST@PLGA nanoparticles (AST@PLGA@BEVs). The preparation process and methods were optimized by three factors and three levels of response surface method to increase drug loading (DL). After optimization, the DL was increased to 6.824%, and the size, polydispersity index, and zeta potential of AST@PLGA@BEVs reached 191.60 ± 2.23 nm, 0.166, and -15.85 ± 0.92 mV, respectively. Moreover, AST@PLGA@BEVs exhibited more notable anticancer activity than AST in vitro. Collectively, these results indicate that the method of loading AST in broccoli-derived EVs is feasible and has important significance for the further development and utilization of AST as a functional food.
Collapse
|
30
|
Wang Y, Cao Z, Wei Q, Ma K, Hu W, Huang Q, Su J, Li H, Zhang C, Fu X. VH298-loaded extracellular vesicles released from gelatin methacryloyl hydrogel facilitate diabetic wound healing by HIF-1α-mediated enhancement of angiogenesis. Acta Biomater 2022; 147:342-355. [PMID: 35580827 DOI: 10.1016/j.actbio.2022.05.018] [Citation(s) in RCA: 88] [Impact Index Per Article: 44.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 04/30/2022] [Accepted: 05/10/2022] [Indexed: 02/07/2023]
Abstract
Endothelial malfunction is responsible for impaired angiogenesis in diabetic patients, thereby causing the delayed healing progress of diabetic wounds. Exosomes or extracellular vesicles (EVs) have emerged as potential therapeutic vectors carrying drug cargoes to diseased cells. In the present study, EVs were reported as a new treatment for diabetic wounds by delivering VH298 into endothelial cells. Firstly, EVs derived from epidermal stem cells (ESCs) were loaded with VH298 (VH-EVs), and the characteristics of VH-EVs were identified. VH-EVs showed promotive action on the function of human umbilical vein endothelial cells (HUVECs) in vitro by activating HIF-1α signaling pathway. VH-EVs were also found to have a therapeutic effect on wound healing and angiogenesis in vivo. We further fabricated gelatin methacryloyl (GelMA) hydrogel for sustained release of VH-EVs, which possessed high biocompatibility and proper mechanical properties. In diabetic mice, GelMA hydrogel containing VH-EVs (Gel-VH-EVs) effectively promoted wound healing by locally enhancing blood supply and angiogenesis. The underlying mechanism for enhanced angiogenesis was possibly associated with the activation of HIF-1α/VEGFA signaling pathway. Collectively, our findings suggest a promising EV-based strategy for the VH298 delivery to endothelial cells and provide a new bioactive dressing for diabetic wound treatment. STATEMENT OF SIGNIFICANCE: The angiogenic dysfunction is the main cause of diabetic wound unhealing. Extracellular vesicles (EVs) have been reported to be helpful but their efficacy is limited for angiogenesis in cutaneous regeneration. VH298 holds great promise to improve angiogenesis by stabilizing HIF-1α which is reported at low level in diabetic wounds. Here, we loaded EVs with VH298 (VH-EVs) to exert an on-target enhancement of proangiogenic capacity in diabetic wound. Then, we applied a photo-crosslinkable hydrogel, gelatin methacryloyl (GelMA) containing VH-EVs (Gel-VH-EVs) as a convenient biomaterial and an adaptable scaffold for sustained releasing VH-EVs. The results showed significant therapeutic effect of Gel-VH-EVs on skin defect repair. Our findings suggest a promising EVs-based drug delivery strategy and a new functional wound dressing for patients.
Collapse
Affiliation(s)
- Yaxi Wang
- Research Center for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research Division and the 4th Medical Center of Chinese PLA General Hospital, Beijing, 100048, China; Chinese PLA Medical School, Beijing, 100853, China
| | - Zhen Cao
- Department of General Surgery, The First Medical Center of Chinese PLA General Hospital, Beijing, 100853, China
| | - Qian Wei
- Research Center for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research Division and the 4th Medical Center of Chinese PLA General Hospital, Beijing, 100048, China
| | - Kui Ma
- Research Center for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research Division and the 4th Medical Center of Chinese PLA General Hospital, Beijing, 100048, China; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing, 100048, China; PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration, Beijing, 100048, China
| | - Wenzhi Hu
- Research Center for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research Division and the 4th Medical Center of Chinese PLA General Hospital, Beijing, 100048, China
| | - Qilin Huang
- Research Center for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research Division and the 4th Medical Center of Chinese PLA General Hospital, Beijing, 100048, China
| | - Jianlong Su
- School of Medicine, NanKai University, Tianjin, 300074, China
| | - Haihong Li
- Department of Wound Repair and Dermatologic Surgery, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000, China.
| | - Cuiping Zhang
- Research Center for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research Division and the 4th Medical Center of Chinese PLA General Hospital, Beijing, 100048, China; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing, 100048, China; PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration, Beijing, 100048, China.
| | - Xiaobing Fu
- Research Center for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research Division and the 4th Medical Center of Chinese PLA General Hospital, Beijing, 100048, China; Chinese PLA Medical School, Beijing, 100853, China; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing, 100048, China; PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration, Beijing, 100048, China.
| |
Collapse
|
31
|
Xiu H, Nan X, Guo D, Wang J, Li J, Peng Y, Xiong G, Wang S, Wang C, Zhang G, Yang Y, Cai Z. Gp350-anchored extracellular vesicles: promising vehicles for delivering therapeutic drugs of B cell malignancies. Asian J Pharm Sci 2022; 17:462-474. [PMID: 35782327 PMCID: PMC9237600 DOI: 10.1016/j.ajps.2022.03.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 02/18/2022] [Accepted: 03/21/2022] [Indexed: 11/27/2022] Open
Abstract
Although chimeric antigen receptor-modified (CAR) T cell therapy has been successfully applied in the treatment of acute B lymphocytic leukemia, its effect on Burkitt lymphoma (BL) and chronic B lymphocytic leukemia (B-CLL) is unsatisfactory. Moreover, fatal side effects greatly impede CAR T cell application. Extracellular vesicles (EVs) are excellent carriers of therapeutic agents. Nevertheless, EVs mainly accumulate in the liver when administered without modification. As an envelope glycoprotein of Epstein-Barr viruses, gp350 can efficiently bind CD21 on B cells. Here, gp350 was directly anchored onto red blood cell EVs (RBC-EVs) via its transmembrane region combined with low-voltage electroporation. The results showed that gp350 could anchor to RBC-EVs with high efficiency and that the resulting gp350-anchored RBC-EVs (RBC-EVs/gp350Etp) exhibited increased targeting to CD21+ BL and B-CLL relative to RBC-EVs. After the loading of doxorubicin or fludarabine, RBC-EVs/gp350Etp had powerful cytotoxicity and therapeutic efficacy on CD21+ BL or B-CLL, respectively. Moreover, RBC-EVs/gp350Etp loaded with a drug did not exhibit any apparent systemic toxicity and specifically induced the apoptosis of tumor B cells but not normal B cells. Therefore, our findings indicate that drug-loaded RBC-EVs/gp350Etp may be adopted in the treatment of CD21+ B cell malignancies.
Collapse
Affiliation(s)
- Huiqing Xiu
- Institute of Immunology, and Department of Orthopedics of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xi Nan
- Institute of Immunology, and Department of Orthopedics of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Danfeng Guo
- Henan Key Laboratory for Digestive Organ Transplantation, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jiaoli Wang
- Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang University Cancer Centre, Hangzhou, China
| | - Jiahui Li
- Department of Critical Care Medicine of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yanmei Peng
- Institute of Immunology, and Department of Orthopedics of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Guirun Xiong
- Department of Emergency Medicine, Tongde Hospital of Zhejiang Province, Hangzhou, China
| | - Shibo Wang
- Institute of Immunology, and Department of Orthopedics of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Changjun Wang
- Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang Provincial Key Lab of Ophthalmology, Hangzhou, China
| | - Gensheng Zhang
- Department of Critical Care Medicine of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yunshan Yang
- Department of Medical Oncology, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Zhijian Cai
- Institute of Immunology, and Department of Orthopedics of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
32
|
Fang Z, Zhang X, Huang H, Wu J. Exosome based miRNA delivery strategy for disease treatment. CHINESE CHEM LETT 2022. [DOI: 10.1016/j.cclet.2021.11.050] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|
33
|
Xu K, Jin Y, Li Y, Huang Y, Zhao R. Recent Progress of Exosome Isolation and Peptide Recognition-Guided Strategies for Exosome Research. Front Chem 2022; 10:844124. [PMID: 35281563 PMCID: PMC8908031 DOI: 10.3389/fchem.2022.844124] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Accepted: 02/14/2022] [Indexed: 12/12/2022] Open
Abstract
Exosomes are membrane extracellular vesicles secreted by almost all kinds of cells, which are rich in proteins, lipids, and nucleic acids. As a medium of intercellular communication, exosomes play important roles in biological processes and are closely related to the occurrence, and development of many diseases. The isolation of exosomes and downstream analyses can provide important information to the accurate diagnosis and treatment of diseases. However, exosomes are various in a size range from 30 to 200 nm and exist in complex bio-systems, which provide significant challenges for the isolation and enrichment of exosomes. Different methods have been developed to isolate exosomes, such as the “gold-standard” ultracentrifugation, size-exclusion chromatography, and polymer precipitation. In order to improve the selectivity of isolation, affinity capture strategies based on molecular recognition are becoming attractive. In this review, we introduced the main strategies for exosome isolation and enrichment, and compared their strengths and limitations. Furthermore, combined with the excellent performance of targeted peptides, we summarized the application of peptide recognition in exosome isolation and engineering modification.
Collapse
Affiliation(s)
- Kun Xu
- Beijing National Laboratory for Molecular Sciences, CAS Key Laboratory of Analytical Chemistry for Living Biosystems, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing, China
- School of Chemistry, University of Chinese Academy of Sciences, Beijing, China
| | - Yulong Jin
- Beijing National Laboratory for Molecular Sciences, CAS Key Laboratory of Analytical Chemistry for Living Biosystems, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing, China
- School of Chemistry, University of Chinese Academy of Sciences, Beijing, China
- *Correspondence: Yulong Jin, ; Rui Zhao,
| | - Yongming Li
- Beijing National Laboratory for Molecular Sciences, CAS Key Laboratory of Analytical Chemistry for Living Biosystems, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing, China
- School of Chemistry, University of Chinese Academy of Sciences, Beijing, China
| | - Yanyan Huang
- Beijing National Laboratory for Molecular Sciences, CAS Key Laboratory of Analytical Chemistry for Living Biosystems, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing, China
- School of Chemistry, University of Chinese Academy of Sciences, Beijing, China
| | - Rui Zhao
- Beijing National Laboratory for Molecular Sciences, CAS Key Laboratory of Analytical Chemistry for Living Biosystems, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing, China
- School of Chemistry, University of Chinese Academy of Sciences, Beijing, China
- *Correspondence: Yulong Jin, ; Rui Zhao,
| |
Collapse
|
34
|
Ostermeier B, Soriano-Sarabia N, Maggirwar SB. Platelet-Released Factors: Their Role in Viral Disease and Applications for Extracellular Vesicle (EV) Therapy. Int J Mol Sci 2022; 23:2321. [PMID: 35216433 PMCID: PMC8876984 DOI: 10.3390/ijms23042321] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 02/13/2022] [Accepted: 02/17/2022] [Indexed: 02/04/2023] Open
Abstract
Platelets, which are small anuclear cell fragments, play important roles in thrombosis and hemostasis, but also actively release factors that can both suppress and induce viral infections. Platelet-released factors include sCD40L, microvesicles (MVs), and alpha granules that have the capacity to exert either pro-inflammatory or anti-inflammatory effects depending on the virus. These factors are prime targets for use in extracellular vesicle (EV)-based therapy due to their ability to reduce viral infections and exert anti-inflammatory effects. While there are some studies regarding platelet microvesicle-based (PMV-based) therapy, there is still much to learn about PMVs before such therapy can be used. This review provides the background necessary to understand the roles of platelet-released factors, how these factors might be useful in PMV-based therapy, and a critical discussion of current knowledge of platelets and their role in viral diseases.
Collapse
Affiliation(s)
| | | | - Sanjay B. Maggirwar
- Department of Microbiology Immunology and Tropical Medicine, The George Washington University, 2300 I Street NW, Washington, DC 20037, USA; (B.O.); (N.S.-S.)
| |
Collapse
|
35
|
Nguyen NP, Helmbrecht H, Ye Z, Adebayo T, Hashi N, Doan MA, Nance E. Brain Tissue-Derived Extracellular Vesicle Mediated Therapy in the Neonatal Ischemic Brain. Int J Mol Sci 2022; 23:620. [PMID: 35054800 PMCID: PMC8775954 DOI: 10.3390/ijms23020620] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 12/23/2021] [Accepted: 12/28/2021] [Indexed: 02/04/2023] Open
Abstract
Hypoxic-Ischemic Encephalopathy (HIE) in the brain is the leading cause of morbidity and mortality in neonates and can lead to irreparable tissue damage and cognition. Thus, investigating key mediators of the HI response to identify points of therapeutic intervention has significant clinical potential. Brain repair after HI requires highly coordinated injury responses mediated by cell-derived extracellular vesicles (EVs). Studies show that stem cell-derived EVs attenuate the injury response in ischemic models by releasing neuroprotective, neurogenic, and anti-inflammatory factors. In contrast to 2D cell cultures, we successfully isolated and characterized EVs from whole brain rat tissue (BEV) to study the therapeutic potential of endogenous EVs. We showed that BEVs decrease cytotoxicity in an ex vivo oxygen glucose deprivation (OGD) brain slice model of HI in a dose- and time-dependent manner. The minimum therapeutic dosage was determined to be 25 μg BEVs with a therapeutic application time window of 4-24 h post-injury. At this therapeutic dosage, BEV treatment increased anti-inflammatory cytokine expression. The morphology of microglia was also observed to shift from an amoeboid, inflammatory phenotype to a restorative, anti-inflammatory phenotype between 24-48 h of BEV exposure after OGD injury, indicating a shift in phenotype following BEV treatment. These results demonstrate the use of OWH brain slices to facilitate understanding of BEV activity and therapeutic potential in complex brain pathologies for treating neurological injury in neonates.
Collapse
Affiliation(s)
- Nam Phuong Nguyen
- Molecular Engineering & Sciences Institute, University of Washington, Seattle, WA 98195, USA;
| | - Hawley Helmbrecht
- Department of Chemical Engineering, University of Washington, Seattle, WA 98195, USA; (H.H.); (Z.Y.); (N.H.)
| | - Ziming Ye
- Department of Chemical Engineering, University of Washington, Seattle, WA 98195, USA; (H.H.); (Z.Y.); (N.H.)
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Tolulope Adebayo
- Department of Biology, University of Washington, Seattle, WA 98195, USA;
| | - Najma Hashi
- Department of Chemical Engineering, University of Washington, Seattle, WA 98195, USA; (H.H.); (Z.Y.); (N.H.)
| | - My-Anh Doan
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA;
| | - Elizabeth Nance
- Molecular Engineering & Sciences Institute, University of Washington, Seattle, WA 98195, USA;
- Department of Chemical Engineering, University of Washington, Seattle, WA 98195, USA; (H.H.); (Z.Y.); (N.H.)
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA;
| |
Collapse
|
36
|
Jordan S, Zielinski M, Kortylewski M, Kuhn T, Bystritsky A. Noninvasive Delivery of Biologicals to the Brain. FOCUS (AMERICAN PSYCHIATRIC PUBLISHING) 2022; 20:64-70. [PMID: 35746928 PMCID: PMC9063603 DOI: 10.1176/appi.focus.20210028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
In the past, psychotherapy and neuropharmacological approaches have been the most common treatments for disordered thoughts, moods, and behaviors. One new path of brain therapeutics is in the deployment of noninvasive approaches designed to reprogram brain function at the cellular level. Treatment at the cellular level may be considered for a wide array of disorders, ranging from mood disorders to neurodegenerative disorders. Brain-targeted biological therapy may provide minimally invasive and accurate delivery of treatment. The present article discusses the hurdles and advances that characterize the pathway to this goal.
Collapse
|
37
|
Bibliography. FOCUS (AMERICAN PSYCHIATRIC PUBLISHING) 2022; 20:76-78. [PMID: 35746926 PMCID: PMC9063592 DOI: 10.1176/appi.focus.20107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
38
|
Hercher D, Nguyen MQ, Dworak H. Extracellular vesicles and their role in peripheral nerve regeneration. Exp Neurol 2021; 350:113968. [PMID: 34973963 DOI: 10.1016/j.expneurol.2021.113968] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 12/04/2021] [Accepted: 12/25/2021] [Indexed: 12/18/2022]
Abstract
Peripheral nerve injuries often result in sensory and motor dysfunction in respective parts of the body. Regeneration after peripheral nerve injuries is a complex process including the differentiation of Schwann cells, recruiting of macrophages, blood vessel growth and axonal regrowth. Extracellular vesicles (EVs) are considered to play a pivotal role in intercellular communication and transfer of biological information. Specifically, their bioactivity and ability to deliver cargos of various types of nucleic acids and proteins have made them a potential vehicle for neurotherapeutics. However, production, characterization, dosage and targeted delivery of EVs still pose challenges for the clinical translation of EV therapeutics. This review summarizes the current knowledge of EVs in the context of the healthy and injured peripheral nerve and addresses novel concepts for modification of EVs as therapeutic agents for peripheral nerve regeneration.
Collapse
Affiliation(s)
- David Hercher
- Ludwig Boltzmann Institute for Traumatology, The Research Center in Cooperation with AUVA, Vienna, Austria; Austrian Cluster for Tissue Regeneration, Vienna, Austria.
| | - Mai Quyen Nguyen
- Ludwig Boltzmann Institute for Traumatology, The Research Center in Cooperation with AUVA, Vienna, Austria; Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Helene Dworak
- Ludwig Boltzmann Institute for Traumatology, The Research Center in Cooperation with AUVA, Vienna, Austria; Ludwig Boltzmann Research Group Senescence and Healing of Wounds, Vienna, Austria; Austrian Cluster for Tissue Regeneration, Vienna, Austria
| |
Collapse
|
39
|
Chen C, Sun M, Wang J, Su L, Lin J, Yan X. Active cargo loading into extracellular vesicles: Highlights the heterogeneous encapsulation behaviour. J Extracell Vesicles 2021; 10:e12163. [PMID: 34719860 PMCID: PMC8558234 DOI: 10.1002/jev2.12163] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Revised: 10/08/2021] [Accepted: 10/13/2021] [Indexed: 12/21/2022] Open
Abstract
Extracellular vesicles (EVs) have demonstrated unique advantages in serving as nanocarriers for drug delivery, yet the cargo encapsulation efficiency is far from expectation, especially for hydrophilic chemotherapeutic drugs. Besides, the intrinsic heterogeneity of EVs renders it difficult to evaluate drug encapsulation behaviour. Inspired by the active drug loading strategy of liposomal nanomedicines, here we report the development of a method, named "Sonication and Extrusion-assisted Active Loading" (SEAL), for effective and stable drug encapsulation of EVs. Using doxorubicin-loaded milk-derived EVs (Dox-mEVs) as the model system, sonication was applied to temporarily permeabilize the membrane, facilitating the influx of ammonium sulfate solution into the lumen to establish the transmembrane ion gradient essential for active loading. Along with extrusion to downsize large mEVs, homogenize particle size and reshape the nonspherical or multilamellar vesicles, SEAL showed around 10-fold enhancement of drug encapsulation efficiency compared with passive loading. Single-particle analysis by nano-flow cytometry was further employed to reveal the heterogeneous encapsulation behaviour of Dox-mEVs which would otherwise be overlooked by bulk-based approaches. Correlation analysis between doxorubicin auto-fluorescence and the fluorescence of a lipophilic dye DiD suggested that only the lipid-enclosed particles were actively loadable. Meanwhile, immunofluorescence analysis revealed that more than 85% of the casein positive particles was doxorubicin free. These findings further inspired the development of the lipid-probe- and immuno-mediated magnetic isolation techniques to selectively remove the contaminants of non-lipid enclosed particles and casein assemblies, respectively. Finally, the intracellular assessments confirmed the superior performance of SEAL-prepared mEV formulations, and demonstrated the impact of encapsulation heterogeneity on therapeutic outcome. The as-developed cargo-loading approach and nano-flow cytometry-based characterization method will provide an instructive insight in the development of EV-based delivery systems.
Collapse
Affiliation(s)
- Chaoxiang Chen
- Department of Biological Engineering, College of Food and Biological EngineeringJimei UniversityXiamenFujianPeople's Republic of China
| | - Mengdi Sun
- Department of Biological Engineering, College of Food and Biological EngineeringJimei UniversityXiamenFujianPeople's Republic of China
| | - Jialin Wang
- Department of Biological Engineering, College of Food and Biological EngineeringJimei UniversityXiamenFujianPeople's Republic of China
| | - Liyun Su
- Department of Chemical Biology, MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, Key Laboratory for Chemical Biology of Fujian Province, Collaborative Innovation Center of Chemistry for Energy Materials, College of Chemistry and Chemical EngineeringXiamen UniversityXiamenFujianPeople's Republic of China
| | - Junjie Lin
- Department of Biological Engineering, College of Food and Biological EngineeringJimei UniversityXiamenFujianPeople's Republic of China
| | - Xiaomei Yan
- Department of Chemical Biology, MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, Key Laboratory for Chemical Biology of Fujian Province, Collaborative Innovation Center of Chemistry for Energy Materials, College of Chemistry and Chemical EngineeringXiamen UniversityXiamenFujianPeople's Republic of China
| |
Collapse
|
40
|
Design and Optimization of the Circulatory Cell-Driven Drug Delivery Platform. Stem Cells Int 2021; 2021:8502021. [PMID: 34603454 PMCID: PMC8481068 DOI: 10.1155/2021/8502021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 08/17/2021] [Indexed: 01/14/2023] Open
Abstract
Achievement of high targeting efficiency for a drug delivery system remains a challenge of tumor diagnoses and nonsurgery therapies. Although nanoparticle-based drug delivery systems have made great progress in extending circulation time, improving durability, and controlling drug release, the targeting efficiency remains low. And the development is limited to reducing side effects since overall survival rates are mostly unchanged. Therefore, great efforts have been made to explore cell-driven drug delivery systems in the tumor area. Cells, particularly those in the blood circulatory system, meet most of the demands that the nanoparticle-based delivery systems do not. These cells possess extended circulation times and innate chemomigration ability and can activate an immune response that exerts therapeutic effects. However, new challenges have emerged, such as payloads, cell function change, cargo leakage, and in situ release. Generally, employing cells from the blood circulatory system as cargo carriers has achieved great benefits and paved the way for tumor diagnosis and therapy. This review specifically covers (a) the properties of red blood cells, monocytes, macrophages, neutrophils, natural killer cells, T lymphocytes, and mesenchymal stem cells; (b) the loading strategies to balance cargo amounts and cell function balance; (c) the cascade strategies to improve cell-driven targeting delivery efficiency; and (d) the features and applications of cell membranes, artificial cells, and extracellular vesicles in cancer treatment.
Collapse
|
41
|
Xie S, Hou X, Yang W, Shi W, Yang X, Duan S, Mo F, Liu A, Wang W, Lu X. Endoglin-Aptamer-Functionalized Liposome-Equipped PD-1-Silenced T Cells Enhance Antitumoral Immunotherapeutic Effects. Int J Nanomedicine 2021; 16:6017-6034. [PMID: 34511903 PMCID: PMC8418331 DOI: 10.2147/ijn.s317220] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 08/11/2021] [Indexed: 12/25/2022] Open
Abstract
Background The broader application of adoptive cell therapy (ACT) in cancer immunotherapies (particularly for solid tumors) has always been limited by the immunosuppressive tumor microenvironment (TME) and the insufficient targetability of effector T cells, resulting in unsatisfied therapeutic outcome. Here, we designed a new strategy by using aptamer-based immunoliposomes to modify PD-1-silencing T cells, which were activated by dendritic cell (DC)/tumor fusion cells (FCs) to improve the antitumor potency of cytotoxic T lymphocytes (CTLs/CD8+ T cells). Methods PD-1 gene was knocked out from CD8+ T cells using CRISPR/Cas9 system to liberate T cell activity from immunosuppression. The PD-1− T cells were stimulated with DC/tumor FCs, followed by further functional modification of tumor-specific nanoliposomes (hEnd-Apt/CD3-Lipo) to generate FC/PD-1− CTLs. The activation and proliferation and specificity of the modified FC/PD-1− CTLs were measured. The antitumor activity of these CTLs against HepG2-tumors was evaluated in xenograft NOD/SCID mice, and the antitumor mechanism was investigated based on tissue immunohistochemistry and serum ELISA. Results Our results indicated that the modification of hEnd-Apt/CD3-Lipo nanocomposites on the FC/PD-1− CTLs had a more substantial synergetic effect in inhibiting tumor growth and prolonging animal survival, rather than other control liposomes. Furthermore, the hEnd-Apt/CD3-Lipo-modified FC/PD-1− CTLs showed a stronger antitumor outcome in the tumor-bearing mouse model, through the mechanisms of suppressing tumor cell proliferation, promoting tumor apoptosis, reducing angiogenesis but increasing the infiltration of the FC/PD-1− CTLs in the tumor tissue, as well as upregulating the systemic levels of IFN-γ, IL-2, TNF-α and IL-6 cytokines, by comparison of the control settings. Conclusion In sum, our investigation suggests an enhancement of antitumor effect by the surface modification of endoglin-targeting nanoliposomes upon DC/tumor FC-activated PD-1− CTLs, therefore, provides a new tumoral endoglin-targeted approach as a promising strategy to reduce immunosuppression of tumor microenvironment and improve the immunotherapeutic outcome of anticancer ACT.
Collapse
Affiliation(s)
- Shenxia Xie
- School of Preclinical Medicine, Guangxi Medical University, Nanning, Guangxi, People's Republic of China.,Pharmaceutical College, Guangxi Medical University, Nanning, Guangxi, People's Republic of China
| | - Xiaoqiong Hou
- School of Preclinical Medicine, Guangxi Medical University, Nanning, Guangxi, People's Republic of China.,International Nanobody Research Center of Guangxi, Guangxi Medical University, Nanning, Guangxi, People's Republic of China
| | - Wei Yang
- School of Preclinical Medicine, Guangxi Medical University, Nanning, Guangxi, People's Republic of China.,International Nanobody Research Center of Guangxi, Guangxi Medical University, Nanning, Guangxi, People's Republic of China
| | - Wei Shi
- School of Preclinical Medicine, Guangxi Medical University, Nanning, Guangxi, People's Republic of China.,International Nanobody Research Center of Guangxi, Guangxi Medical University, Nanning, Guangxi, People's Republic of China
| | - Xiaomei Yang
- School of Preclinical Medicine, Guangxi Medical University, Nanning, Guangxi, People's Republic of China.,International Nanobody Research Center of Guangxi, Guangxi Medical University, Nanning, Guangxi, People's Republic of China
| | - Siliang Duan
- International Nanobody Research Center of Guangxi, Guangxi Medical University, Nanning, Guangxi, People's Republic of China
| | - Fengzhen Mo
- International Nanobody Research Center of Guangxi, Guangxi Medical University, Nanning, Guangxi, People's Republic of China
| | - Aiqun Liu
- International Nanobody Research Center of Guangxi, Guangxi Medical University, Nanning, Guangxi, People's Republic of China
| | - Wu Wang
- School of Preclinical Medicine, Guangxi Medical University, Nanning, Guangxi, People's Republic of China.,Laboratory of Tropical Biomedicine and Biotechnology, School of Tropical Medicine and Laboratory Medicine, Hainan Medical University, Haikou, Hainan, 571101, People's Republic of China
| | - Xiaoling Lu
- School of Preclinical Medicine, Guangxi Medical University, Nanning, Guangxi, People's Republic of China.,International Nanobody Research Center of Guangxi, Guangxi Medical University, Nanning, Guangxi, People's Republic of China.,College of Stomatology, Guangxi Medical University, Nanning, Guangxi, People's Republic of China
| |
Collapse
|
42
|
Escudé Martinez de Castilla P, Tong L, Huang C, Sofias AM, Pastorin G, Chen X, Storm G, Schiffelers RM, Wang JW. Extracellular vesicles as a drug delivery system: A systematic review of preclinical studies. Adv Drug Deliv Rev 2021; 175:113801. [PMID: 34015418 DOI: 10.1016/j.addr.2021.05.011] [Citation(s) in RCA: 90] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 05/10/2021] [Accepted: 05/15/2021] [Indexed: 02/06/2023]
Abstract
During the past decades, extracellular vesicles (EVs) have emerged as an attractive drug delivery system. Here, we assess their pre-clinical applications, in the form of a systematic review. For each study published in the past decade, disease models, animal species, EV donor cell types, active pharmaceutical ingredients (APIs), EV surface modifications, API loading methods, EV size and charge, estimation of EV purity, presence of biodistribution studies and administration routes were quantitatively analyzed in a defined and reproducible way. We have interpreted the trends we observe over the past decade, to define the niches where to apply EVs for drug delivery in the future and to provide a basis for regulatory guidelines.
Collapse
|
43
|
Xun J, Li C, Liu M, Mei Y, Zhou Q, Wu B, Xie F, Liu Y, Dai R. Serum exosomes from young rats improve the reduced osteogenic differentiation of BMSCs in aged rats with osteoporosis after fatigue loading in vivo. Stem Cell Res Ther 2021; 12:424. [PMID: 34315544 PMCID: PMC8314589 DOI: 10.1186/s13287-021-02449-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 06/06/2021] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Osteoporosis is a major public health concern for the elderly population and is characterized by fatigue load resulting in bone microdamage. The ability of bone mesenchymal stem cells (BMSCs) to repair bone microdamage diminishes with age, and the accumulation of bone microdamage increases the risk of osteoporotic fracture. There is a lack of effective means to promote the repair of bone microdamage in aged patients with osteoporosis. Exosomes have been shown to be related to the osteogenic differentiation of BMSCs. Here, we aimed to evaluate the changes in the osteogenic differentiation capacity of BMSCs in aged osteoporotic rats after fatigue loading and the treatment potential of serum exosomes from young rats. METHODS The tibias of six aged osteoporotic rats were subjected to fatigue loading in vivo for 2 weeks, and the bone microdamage, microstructures, and mechanical properties were assessed. Subsequently, BMSCs were extracted to evaluate their proliferation and osteogenic differentiation abilities. In addition, the BMSCs of aged osteoporotic rats after fatigue loading were treated with serum exosomes from young rats under osteogenic induction conditions, and the expression of osteogenic-related miRNAs was quantified. The osteogenetic effects of miRNA-19b-3p in exosomes and the possible target protein PTEN was detected. RESULTS Obvious bone microdamage at the fatigue load stress point, the bone microstructure and biomechanical properties were not obviously changed. A decreased osteogenic differentiation ability of BMSCs was observed after fatigue loading, while serum exosomes from young rats highly expressing miRNA-19b-3p improved the decreased osteogenic differentiation ability of BMSCs. Transfection with miRNA-19b-3p mimic could promote osteoblastic differentiation of BMSCs and decreased the expression of PTEN. After transfection of miRNA-19b-3p inhibitor, the promotional effect of exosomes on bone differentiation was weakened. Treatment with transfected exosomes increased the expression of PTEN. CONCLUSION Serum exosomes derived from young rats can improve the decreased osteogenic differentiation ability of BMSCs in aged rats with osteoporosis after fatigue loading and can provide a new treatment strategy for the repair of bone microdamage and prevention of fractures.
Collapse
Affiliation(s)
- Jingqiong Xun
- National Clinical Research Center for Metabolic Diseases, Institute of Metabolism and Endocrinology, Central South University, Hunan Provincial Key Laboratory of Metabolic Bone Diseases, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Department of Endocrinology, Affiliated Hospital of Guilin Medical University, Guilin, Guangxi, China
| | - Chan Li
- National Clinical Research Center for Metabolic Diseases, Institute of Metabolism and Endocrinology, Central South University, Hunan Provincial Key Laboratory of Metabolic Bone Diseases, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Meilu Liu
- National Clinical Research Center for Metabolic Diseases, Institute of Metabolism and Endocrinology, Central South University, Hunan Provincial Key Laboratory of Metabolic Bone Diseases, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Yueming Mei
- National Clinical Research Center for Metabolic Diseases, Institute of Metabolism and Endocrinology, Central South University, Hunan Provincial Key Laboratory of Metabolic Bone Diseases, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Qiongfei Zhou
- National Clinical Research Center for Metabolic Diseases, Institute of Metabolism and Endocrinology, Central South University, Hunan Provincial Key Laboratory of Metabolic Bone Diseases, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Bo Wu
- National Clinical Research Center for Metabolic Diseases, Institute of Metabolism and Endocrinology, Central South University, Hunan Provincial Key Laboratory of Metabolic Bone Diseases, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Fen Xie
- Department of Endocrinology, Xiangtan Central Hospital, Xiangtan, Hunan, China
| | - Yuling Liu
- National Clinical Research Center for Metabolic Diseases, Institute of Metabolism and Endocrinology, Central South University, Hunan Provincial Key Laboratory of Metabolic Bone Diseases, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Ruchun Dai
- National Clinical Research Center for Metabolic Diseases, Institute of Metabolism and Endocrinology, Central South University, Hunan Provincial Key Laboratory of Metabolic Bone Diseases, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China.
| |
Collapse
|
44
|
Chen L, Hong W, Ren W, Xu T, Qian Z, He Z. Recent progress in targeted delivery vectors based on biomimetic nanoparticles. Signal Transduct Target Ther 2021; 6:225. [PMID: 34099630 PMCID: PMC8182741 DOI: 10.1038/s41392-021-00631-2] [Citation(s) in RCA: 112] [Impact Index Per Article: 37.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Revised: 04/28/2021] [Accepted: 04/29/2021] [Indexed: 02/05/2023] Open
Abstract
Over the past decades, great interest has been given to biomimetic nanoparticles (BNPs) since the rise of targeted drug delivery systems and biomimetic nanotechnology. Biological vectors including cell membranes, extracellular vesicles (EVs), and viruses are considered promising candidates for targeted delivery owing to their biocompatibility and biodegradability. BNPs, the integration of biological vectors and functional agents, are anticipated to load cargos or camouflage synthetic nanoparticles to achieve targeted delivery. Despite their excellent intrinsic properties, natural vectors are deliberately modified to endow multiple functions such as good permeability, improved loading capability, and high specificity. Through structural modification and transformation of the vectors, they are pervasively utilized as more effective vehicles that can deliver contrast agents, chemotherapy drugs, nucleic acids, and genes to target sites for refractory disease therapy. This review summarizes recent advances in targeted delivery vectors based on cell membranes, EVs, and viruses, highlighting the potential applications of BNPs in the fields of biomedical imaging and therapy industry, as well as discussing the possibility of clinical translation and exploitation trend of these BNPs.
Collapse
Affiliation(s)
- Li Chen
- Department of Pharmacy, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Weiqi Hong
- Department of Pharmacy, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Wenyan Ren
- Department of Pharmacy, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Ting Xu
- Department of Pharmacy, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, Sichuan, China
| | - Zhiyong Qian
- Department of Pharmacy, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zhiyao He
- Department of Pharmacy, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
45
|
Simonsen JB, Kromann EB. Pitfalls and opportunities in quantitative fluorescence-based nanomedicine studies - A commentary. J Control Release 2021; 335:660-667. [PMID: 34089794 DOI: 10.1016/j.jconrel.2021.05.041] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 05/27/2021] [Accepted: 05/28/2021] [Indexed: 12/24/2022]
Abstract
Fluorescence-based techniques are prevalent in studies of nanomedicine-targeting to cells and tissues. However, fluorescence-based studies are rarely quantitative, thus prohibiting direct comparisons of nanomedicine-performance across studies. With this Commentary, we aim to provoke critical thinking about experimental design by treating some often-overlooked pitfalls in 'quantitative' fluorescence-based experimentation. Focusing on fluorescence-labeled nanoparticles, we cover mechanisms like solvent-interactions and fluorophore-dissociation, which disqualify the assumption that 'a higher fluorescence readout' translates directly to 'a better targeting efficacy'. With departure in recent literature, we propose guidelines for circumventing these pitfalls in studies of tissue-accumulation and cell-uptake, thus covering fluorescence-based techniques like bulk solution fluorescence measurements, fluorescence microscopy, flow cytometry, and infrared fluorescence imaging. With this, we hope to lay a foundation for more 'quantitative thinking' during experimental design, enabling (for example) the estimation and reporting of actual numbers of fluorescent nanoparticles accumulated in cells and organs.
Collapse
Affiliation(s)
- Jens B Simonsen
- Department of Health Technology, Section for Biotherapeutic Engineering and Drug Targeting, Technical University of Denmark, Ørsteds Plads 345C, 2800 Kgs. Lyngby, Denmark.
| | - Emil B Kromann
- Department of Health Technology, Section for Biomimetics, Technical University of Denmark, Ørsteds Plads 345C, 2800 Kgs. Lyngby, Denmark
| |
Collapse
|
46
|
Richter M, Vader P, Fuhrmann G. Approaches to surface engineering of extracellular vesicles. Adv Drug Deliv Rev 2021; 173:416-426. [PMID: 33831479 DOI: 10.1016/j.addr.2021.03.020] [Citation(s) in RCA: 90] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 03/03/2021] [Accepted: 03/29/2021] [Indexed: 12/23/2022]
Abstract
Extracellular vesicles (EVs) are cell-derived nanoparticles that are important mediators in intercellular communication. This function makes them auspicious candidates for therapeutic and drug-delivery applications. Among EVs, mammalian cell derived EVs and outer membrane vesicles (OMVs) produced by gram-negative bacteria are the most investigated candidates for pharmaceutical applications. To further optimize their performance and to utilize their natural abilities, researchers have strived to equip EVs with new moieties on their surface while preserving the integrity of the vesicles. The aim of this review is to give a comprehensive overview of techniques that can be used to introduce these moieties to the vesicle surface. Approaches can be classified in regards to whether they take place before or after the isolation of EVs. The producing cells can be subjected to genetic manipulation or metabolic engineering to produce surface modified vesicles or EVs are engineered after their isolation by physical or chemical means. Here, the advantages and disadvantages of these processes and their applicability for the development of EVs as therapeutic agents are discussed.
Collapse
|
47
|
Hwang HS, Kim H, Han G, Lee JW, Kim K, Kwon IC, Yang Y, Kim SH. Extracellular Vesicles as Potential Therapeutics for Inflammatory Diseases. Int J Mol Sci 2021; 22:5487. [PMID: 34067503 PMCID: PMC8196952 DOI: 10.3390/ijms22115487] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Accepted: 05/17/2021] [Indexed: 12/11/2022] Open
Abstract
Extracellular vesicles (EV) deliver cargoes such as nucleic acids, proteins, and lipids between cells and serve as an intercellular communicator. As it is revealed that most of the functions associated to EVs are closely related to the immune response, the important role of EVs in inflammatory diseases is emerging. EVs can be functionalized through EV surface engineering and endow targeting moiety that allows for the target specificity for therapeutic applications in inflammatory diseases. Moreover, engineered EVs are considered as promising nanoparticles to develop personalized therapeutic carriers. In this review, we highlight the role of EVs in various inflammatory diseases, the application of EV as anti-inflammatory therapeutics, and the current state of the art in EV engineering techniques.
Collapse
Affiliation(s)
- Hee Sook Hwang
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea; (H.S.H.); (H.K.); (G.H.); (J.W.L.); (K.K.); (I.C.K.)
- Department of Pharmaceutical Engineering, Dankook University, Cheonan 31116, Korea
| | - Hyosuk Kim
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea; (H.S.H.); (H.K.); (G.H.); (J.W.L.); (K.K.); (I.C.K.)
| | - Geonhee Han
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea; (H.S.H.); (H.K.); (G.H.); (J.W.L.); (K.K.); (I.C.K.)
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Korea
| | - Jong Won Lee
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea; (H.S.H.); (H.K.); (G.H.); (J.W.L.); (K.K.); (I.C.K.)
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Korea
| | - Kwangmeyung Kim
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea; (H.S.H.); (H.K.); (G.H.); (J.W.L.); (K.K.); (I.C.K.)
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Korea
| | - Ick Chan Kwon
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea; (H.S.H.); (H.K.); (G.H.); (J.W.L.); (K.K.); (I.C.K.)
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Korea
- Department of Cancer Biology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA 02215, USA
| | - Yoosoo Yang
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea; (H.S.H.); (H.K.); (G.H.); (J.W.L.); (K.K.); (I.C.K.)
| | - Sun Hwa Kim
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea; (H.S.H.); (H.K.); (G.H.); (J.W.L.); (K.K.); (I.C.K.)
| |
Collapse
|
48
|
Samal S, Dash P, Dash M. Drug Delivery to the Bone Microenvironment Mediated by Exosomes: An Axiom or Enigma. Int J Nanomedicine 2021; 16:3509-3540. [PMID: 34045855 PMCID: PMC8149288 DOI: 10.2147/ijn.s307843] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 03/30/2021] [Indexed: 12/12/2022] Open
Abstract
The increasing incidence of bone-related disorders is causing a burden on the clinical scenario. Even though bone is one of the tissues that possess tremendous regenerative potential, certain bone anomalies need therapeutic intervention through appropriate delivery of a drug. Among several nanosystems and biologics that offer the potential to contribute towards bone healing, the exosomes from the class of extracellular vesicles are outstanding. Exosomes are extracellular nanovesicles that, apart from the various advantages, are standing out of the crowd for their ability to conduct cellular communication. The internal cargo of the exosomes is leading to its potential use in therapeutics. Exosomes are being unraveled in terms of the mechanism as well as application in targeting various diseases and tissues. Through this review, we have tried to understand and review all that is already established and the gap areas that still exist in utilizing them as drug delivery vehicles targeting the bone. The review highlights the potential of the exosomes towards their contribution to the drug delivery scenario in the bone microenvironment. A comparison of the pros and cons of exosomes with other prevalent drug delivery systems is also done. A section on the patents that have been generated so far from this field is included.
Collapse
Affiliation(s)
- Sasmita Samal
- Institute of Life Sciences, Nalco Square, Bhubaneswar, Odisha, 751023, India
- School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT) University, Bhubaneswar, Odisha, 751024, India
| | - Pratigyan Dash
- Institute of Life Sciences, Nalco Square, Bhubaneswar, Odisha, 751023, India
- School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT) University, Bhubaneswar, Odisha, 751024, India
| | - Mamoni Dash
- Institute of Life Sciences, Nalco Square, Bhubaneswar, Odisha, 751023, India
| |
Collapse
|
49
|
Huang Z, Guo L, Huang L, Shi Y, Liang J, Zhao L. Baicalin-loaded macrophage-derived exosomes ameliorate ischemic brain injury via the antioxidative pathway. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 126:112123. [PMID: 34082940 DOI: 10.1016/j.msec.2021.112123] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 03/30/2021] [Accepted: 04/18/2021] [Indexed: 12/13/2022]
Abstract
Baicalin (BA), a strong free radical scavenger, has been demonstrated to exert neuroprotective effects in the treatment of ischemic stroke. However, its clinical application has been limited due to its inability to target the brain and its poor solubility. In this study, we designed novel brain-targeted BA-loaded macrophage-derived exosomes (Exo-BA) to induce neuroprotection against ischemic stroke in animal models. The results revealed that with the help of Exo, the solubility of BA was significantly enhanced. In addition, Exo-BA displayed better brain targeting ability than free BA, as they induced the transfer of more BA into the brain, in a transient middle cerebral artery occlusion/reperfusion (tMCAO) model and permanent middle cerebral artery occlusion (pMCAO) model. Compared with free BA, Exo-BA significantly reduced the generation of reactive oxygen species (ROS) and activated the Nrf2/HO-1 pathway in neurons, thus significantly alleviating cerebral ischemic injury in a stroke model.
Collapse
Affiliation(s)
- Zhixuan Huang
- School of Pharmacy, Jinzhou Medical University, Jinzhou 121000, PR China
| | - Lin Guo
- School of Pharmacy, Jinzhou Medical University, Jinzhou 121000, PR China
| | - Lijuan Huang
- School of Pharmacy, Jinzhou Medical University, Jinzhou 121000, PR China
| | - Yijie Shi
- School of Pharmacy, Jinzhou Medical University, Jinzhou 121000, PR China.
| | - Jia Liang
- Life Science Institution, Jinzhou Medical University, Jinzhou 121000, PR China.
| | - Liang Zhao
- School of Pharmacy, Jinzhou Medical University, Jinzhou 121000, PR China.
| |
Collapse
|
50
|
Komuro H, Kawai-Harada Y, Aminova S, Pascual N, Malik A, Contag CH, Harada M. Engineering Extracellular Vesicles to Target Pancreatic Tissue In Vivo. Nanotheranostics 2021; 5:378-390. [PMID: 33912378 PMCID: PMC8077969 DOI: 10.7150/ntno.54879] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 03/31/2021] [Indexed: 01/04/2023] Open
Abstract
Extracellular vesicles (EVs) are naturally released, cell-derived vesicles that mediate intracellular communication, in part, by transferring genetic information and, thus, have the potential to be modified for use as a therapeutic gene or drug delivery vehicle. Advances in EV engineering suggest that directed delivery can be accomplished via surface alterations. Here we assess enriched delivery of engineered EVs displaying an organ targeting peptide specific to the pancreas. We first characterized the size, morphology, and surface markers of engineered EVs that were decorated with a recombinant protein specific to pancreatic β-cells. This β-cell-specific recombinant protein consists of the peptide p88 fused to the EV-binding domain of lactadherin (C1C2). These engineered EVs, p88-EVs, specifically bound to pancreatic β-cells in culture and transferred encapsulated plasmid DNA (pDNA) as early as in 10 min suggesting that the internalization of peptide-bearing EVs is a rapid process. Biodistribution of p88-EVs administrated intravenously into mice showed an altered pattern of EV localization and improved DNA delivery to the pancreas relative to control EVs, as well as an accumulation of targeting EVs to the pancreas using luciferase activity as a readout. These findings demonstrate that systemic administration of engineered EVs can efficiently deliver their cargo as gene carriers to targeted organs in live animals.
Collapse
Affiliation(s)
- Hiroaki Komuro
- Department of Cardiovascular Medicine, Tokyo Medical and Dental University, Tokyo, Japan
- Institute for Quantitative Health Science and Engineering (IQ), Michigan State University, Michigan, USA
- Department of Biomedical Engineering, Michigan State University, Michigan, USA
| | - Yuki Kawai-Harada
- Institute for Quantitative Health Science and Engineering (IQ), Michigan State University, Michigan, USA
- Department of Biomedical Engineering, Michigan State University, Michigan, USA
| | - Shakhlo Aminova
- Institute for Quantitative Health Science and Engineering (IQ), Michigan State University, Michigan, USA
- Lyman Briggs College, Michigan State University, Michigan, USA
| | - Nathaniel Pascual
- Institute for Quantitative Health Science and Engineering (IQ), Michigan State University, Michigan, USA
- Department of Chemical Engineering and Material, Michigan State University, Michigan, USA
| | - Anshu Malik
- Institute for Quantitative Health Science and Engineering (IQ), Michigan State University, Michigan, USA
- Department of Biomedical Engineering, Michigan State University, Michigan, USA
| | - Christopher H. Contag
- Institute for Quantitative Health Science and Engineering (IQ), Michigan State University, Michigan, USA
- Department of Microbiology and Molecular Genetics, Michigan State University, Michigan, USA
- Department of Biomedical Engineering, Michigan State University, Michigan, USA
| | - Masako Harada
- Institute for Quantitative Health Science and Engineering (IQ), Michigan State University, Michigan, USA
- Department of Biomedical Engineering, Michigan State University, Michigan, USA
| |
Collapse
|