1
|
Tang Z, Chen C, Zhou C, Liu Z, Li T, Zhang Y, Feng Y, Gu C, Li S, Chen J. Insights into tumor-derived exosome inhibition in cancer therapy. Eur J Med Chem 2025; 285:117278. [PMID: 39823808 DOI: 10.1016/j.ejmech.2025.117278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 01/11/2025] [Accepted: 01/11/2025] [Indexed: 01/20/2025]
Abstract
Exosomes are critical mediators of cell-to-cell communication in physiological and pathological processes, due to their ability to deliver a variety of bioactive molecules. Tumor-derived exosomes (TDEs), in particular, carry carcinogenic molecules that contribute to tumor progression, metastasis, immune escape, and drug resistance. Thus, TDE inhibition has emerged as a promising strategy to combat cancer. In this review, we discuss the key mechanisms of TDE biogenesis and secretion, emphasizing their implications in tumorigenesis and cancer progression. Moreover, we provide an overview of small-molecule TDE inhibitors that target specific biogenesis and/or secretion pathways, highlighting their potential use in cancer treatment. Lastly, we present the existing obstacles and propose corresponding remedies for the future development of TDE inhibitors.
Collapse
Affiliation(s)
- Ziwei Tang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Cheng Chen
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Chen Zhou
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL, 32610, United States
| | - Zhouyan Liu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Tong Li
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Ye Zhang
- School of Petrochemical Engineering, Changzhou University, Changzhou, 213164, China.
| | - Yanyan Feng
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Chenglei Gu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Shijia Li
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Jichao Chen
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| |
Collapse
|
2
|
Abbasi R, Alamdari-Mahd G, Maleki-Kakelar H, Momen-Mesgin R, Ahmadi M, Sharafkhani M, Rezaie J. Recent advances in the application of engineered exosomes from mesenchymal stem cells for regenerative medicine. Eur J Pharmacol 2025; 989:177236. [PMID: 39753159 DOI: 10.1016/j.ejphar.2024.177236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 12/14/2024] [Accepted: 12/23/2024] [Indexed: 01/12/2025]
Abstract
Exosomes, cell-derived vesicles produced by cells, are fascinating and drawing growing interest in biomedical exploration due to their exceptional properties. There is intriguing evidence that exosomes are involved in major biological processes, including diseases and regeneration. Exosomes from mesenchymal stem cells (MSCs) have shown promising outcomes in regenerative medicine. Numerous studies suggest that exosomes have several advantages over conventional synthetic nanocarriers, opening novel frontiers for innovative drug delivery. Regenerative medicine has demonstrated the profound therapeutic outcomes of engineered or loaded exosomes from MSCs. Different methods are being used to modify or/load exosomes. These exosomes can improve cell signaling pathways for bone and cartilage diseases, liver diseases, nerve tissues, kidney diseases, skin tissue, and cardiovascular diseases. Despite extensive research, clinical translation of these exosomes remains a challenge. The optimization of cargo loading methods, efficiency, physiological stability, and the isolation and characterization of exosomes present some challenges. The upcoming examination should include the development of large-scale, quality-controllable production approaches, the modification of drug loading approaches, and numerous in vivo investigations and clinical trials. Here, we provided an informative overview of the extracellular vesicles and modification/loading methods of exosomes. We discuss the last exosome research on regeneration disorders, highlighting the therapeutic applications of MSCs-derived exosomes. We also highlight future directions and challenges, underscoring the significance of addressing the main questions in the field.
Collapse
Affiliation(s)
- Reza Abbasi
- Department of Biology, Urmia University, Urmia, Iran
| | - Ghazal Alamdari-Mahd
- Solid Tumor Research Center, Cellular and Molecular Medicine Research Institute, Urmia University of Medical Sciences, Urmia, Iran
| | - Hadi Maleki-Kakelar
- Solid Tumor Research Center, Cellular and Molecular Medicine Research Institute, Urmia University of Medical Sciences, Urmia, Iran.
| | | | - Mahdi Ahmadi
- Tuberculosis and Lung Disease Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohaddeseh Sharafkhani
- Solid Tumor Research Center, Cellular and Molecular Medicine Research Institute, Urmia University of Medical Sciences, Urmia, Iran
| | - Jafar Rezaie
- Solid Tumor Research Center, Cellular and Molecular Medicine Research Institute, Urmia University of Medical Sciences, Urmia, Iran.
| |
Collapse
|
3
|
Qian L, Chen P, Zhang S, Wang Z, Guo Y, Koutouratsas V, Fleishman JS, Huang C, Zhang S. The uptake of extracellular vesicles: Research progress in cancer drug resistance and beyond. Drug Resist Updat 2025; 79:101209. [PMID: 39893749 DOI: 10.1016/j.drup.2025.101209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 01/22/2025] [Accepted: 01/26/2025] [Indexed: 02/04/2025]
Abstract
Extracellular vesicles (EVs) are heterogeneous vesicles released by donor cells that can be taken up by recipient cells, thus inducing cellular phenotype changes. Since their discovery decades ago, roles of EVs in modulating initiation, growth, survival and metastasis of cancer have been revealed. Recent studies from multifaceted perspectives have further detailed the contribution of EVs to cancer drug resistance; however, the role of EV uptake in conferring drug resistance seems to be overlooked. In this comprehensive review, we update the EV subtypes and approaches for determining EV uptake. The biological basis of EV uptake is systematically summarized. Moreover, we focus on the diverse uptake mechanisms by which EVs carry out the intracellular delivery of functional molecules and drug resistance signaling. Furthermore, we highlight how EV uptake confers drug resistance and identify potential strategies for targeting EV uptake to overcome drug resistance. Finally, we discuss the research gap on the role of EV uptake in promoting drug resistance. This updated knowledge provides a new avenue to overcome cancer drug resistance by targeting EV uptake.
Collapse
Affiliation(s)
- Luomeng Qian
- Department of Cell Biology, School of Medicine, Nankai University, Tianjin, 300071, China
| | - Pangzhou Chen
- Department of Breast Surgery, Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Foshan 528200, China
| | - Shiwu Zhang
- Department of Pathology, Tianjin Union Medical Center, Nankai University, Tianjin 300121, China
| | - Zhenglu Wang
- Department of Pathology, Tianjin Key Laboratory for Organ Transplantation, Tianjin First Centre Hospital, Tianjin 300192, China
| | - Yuan Guo
- Department of Cell Biology, School of Medicine, Nankai University, Tianjin, 300071, China
| | - Vasili Koutouratsas
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, USA
| | - Joshua S Fleishman
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, USA
| | - Chuanqiang Huang
- Department of Breast Surgery, Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Foshan 528200, China
| | - Sihe Zhang
- Department of Cell Biology, School of Medicine, Nankai University, Tianjin, 300071, China.
| |
Collapse
|
4
|
Yadav K, Sahu KK, Sucheta, Minz S, Pradhan M. Unlocking exosome therapeutics: The critical role of pharmacokinetics in clinical applications. Tissue Cell 2025; 93:102749. [PMID: 39904192 DOI: 10.1016/j.tice.2025.102749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 01/10/2025] [Accepted: 01/15/2025] [Indexed: 02/06/2025]
Abstract
Exosomes are microscopic vesicles released by cells that transport various biological materials and play a vital role in intercellular communication. When they are engineered, they serve as efficient delivery systems for therapeutic agents, making it possible to precisely deliver active pharmaceutical ingredients to organs, tissues, and cells. Exosomes' pharmacokinetics, or how they are transported and metabolized inside the body, is affected by several factors, including their source of origination and the proteins in their cell membranes. The pharmacokinetics and mobility of both native and modified exosomes are being observed in living organisms using advanced imaging modalities such as in vitro-in vivo simulation, magnetic resonance imaging, and positron emission tomography. Establishing comprehensive criteria for the investigation of exosomal pharmacokinetic is essential, given its increasing significance in both therapy and diagnostics. To obtain a thorough understanding of exosome intake, distribution, metabolism, and excretion, molecular imaging methods are crucial. The development of industrial processes and therapeutic applications depends on the precise measurement of exosome concentration in biological samples. To ensure a seamless incorporation of exosomes into clinical practice, as their role in therapeutics grows, it is imperative to conduct a complete assessment of their pharmacokinetics. This review provides a brief on how exosome-based research is evolving and the need for pharmacokinetic consideration to realize the full potential of these promising new therapeutic approaches.
Collapse
Affiliation(s)
- Krishna Yadav
- Rungta College of Pharmaceutical Sciences and Research, Kohka Road, Kurud, Bhilai, Chhattisgarh 491024, India
| | - Kantrol Kumar Sahu
- Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh 281406, India
| | - Sucheta
- School of Medical and Allied Sciences, K. R. Mangalam University, Gurugram, Haryana 11 122103, India
| | - Sunita Minz
- Department of Pharmacy, Indira Gandhi National Tribal University, Amarkantak, India
| | | |
Collapse
|
5
|
Wu J, Tang J, Zhang L, Wang W, Li Z, Zhou L, Jiang X, Huang Y, Guo Q, Wang W, Ding Z, Cai F, Xi K, Gu Y, Chen L. Biomimetic "Trojan Horse" Fibers Modulate Innate Immunity Cascades for Nerve Regeneration. ACS NANO 2025; 19:781-802. [PMID: 39708371 PMCID: PMC11752508 DOI: 10.1021/acsnano.4c12036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 12/07/2024] [Accepted: 12/13/2024] [Indexed: 12/23/2024]
Abstract
Neutrophil membrane vesicles (NMVs) have been successfully applied to control the inflammatory cascade after spinal cord injury (SCI) by acting as an inflammatory factor decoy to front-load the overall inflammation regulatory window; however, the mechanisms by which NMVs regulate macrophage phenotypic shifts as well as their outcomes have rarely been reported. In this study, we demonstrated the "efferocytosis-like" effect of NMVs endocytosed by macrophages, supplementing the TCA cycle intermediate metabolite α-KG by promoting glutamine metabolism, which in turn facilitates oxidative phosphorylation and inhibits the NF-κB signaling pathway to reprogram inflammatory macrophages to the pro-regenerative phenotype. Based on these findings, a "Trojan horse" composite fiber scaffold was constructed; this comprised a carboxylated poly-l-lactic acid shell encapsulated with NMVs and a core loaded with brain-derived neurotrophic factor to spatiotemporally modulate the inflammatory microenvironment by 39.23% and sustainably promote nerve regeneration by 85.67%. In vivo experiments further confirmed the effect of NMV-coated fiber scaffolds on the regulation of early innate immune inflammation and the continuous promotion of nerve regeneration. This study not only further unravels the mechanism of neutrophil membrane-macrophage interactions but also provides a strategy for coordinating inflammatory reprogramming and nerve regeneration following SCI.
Collapse
Affiliation(s)
| | | | | | | | - Ziang Li
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 899 Pinghai Road, Suzhou 215006, Jiangsu, China
| | - Liang Zhou
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 899 Pinghai Road, Suzhou 215006, Jiangsu, China
| | - Xinzhao Jiang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 899 Pinghai Road, Suzhou 215006, Jiangsu, China
| | - Yiyang Huang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 899 Pinghai Road, Suzhou 215006, Jiangsu, China
| | - Qiangqiang Guo
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 899 Pinghai Road, Suzhou 215006, Jiangsu, China
| | - Wenbo Wang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 899 Pinghai Road, Suzhou 215006, Jiangsu, China
| | - Zhouye Ding
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 899 Pinghai Road, Suzhou 215006, Jiangsu, China
| | - Feng Cai
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 899 Pinghai Road, Suzhou 215006, Jiangsu, China
| | - Kun Xi
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 899 Pinghai Road, Suzhou 215006, Jiangsu, China
| | - Yong Gu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 899 Pinghai Road, Suzhou 215006, Jiangsu, China
| | - Liang Chen
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 899 Pinghai Road, Suzhou 215006, Jiangsu, China
| |
Collapse
|
6
|
Chuang CF, Lin CW, Yeh CK. Ultrasound-triggered drug release and cytotoxicity of microbubbles with diverse drug attributes. ULTRASONICS SONOCHEMISTRY 2025; 112:107182. [PMID: 39631357 PMCID: PMC11655813 DOI: 10.1016/j.ultsonch.2024.107182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Accepted: 11/28/2024] [Indexed: 12/07/2024]
Abstract
Ultrasound (US)-triggered cavitation of drug-loaded microbubbles (MBs) represents a promising approach for targeted drug delivery, with substantial benefits attainable through precise control over drug release dosage and form. This study investigates Camptothecin-loaded MBs (CPT-MBs) and Doxorubicin-loaded MBs (DOX-MBs), focusing on how properties such as hydrophilicity, hydrophobicity, and charged functional groups affect their interaction with the lipid surfaces of MBs, thereby influencing the fundamental characteristics and acoustic properties of the drug-loaded MBs. In comparison to DOX-MBs, CPT-MBs showed larger MB size (2.2 ± 0.3 and 1.4 ± 0.1 μm, respectively), a 2-fold increase in drug loading, and an 18 % reduction in leakage after 2 h at 37℃. Under 1 MHz US with a 100 ms pulse repetition interval (PRI), 1000 cycles, 5-minute duration, and 550 kPa acoustic pressure, CPT-MBs undergo inertial cavitation, while DOX-MBs undergo stable cavitation. Drug particles released from these MBs under US-induced cavitation were analyzed using dynamic light scattering, NanoSight, cryo-electron microscopy, and density gradient ultracentrifugation. Results showed that CPT-MBs mainly release free CPT, while DOX-MBs release multilayered DOX-lipid aggregates. The cytotoxicity to C6 cells induced by US-triggered cavitation of these two types of MBs also differed. DOX-lipid aggregates delayed initial uptake, leading to less pronounced short-term (2 h) effects compared to the rapid release of free CPT from CPT-MBs. These findings underscore the need to optimize drug delivery strategies by fine-tuning MB composition and US parameters to control drug release kinetics and achieve the best tumoricidal outcomes.
Collapse
Affiliation(s)
- Chi-Fen Chuang
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan
| | - Chia-Wei Lin
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan
| | - Chih-Kuang Yeh
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan.
| |
Collapse
|
7
|
Gowtham A, Kaundal RK. Exploring the ncRNA landscape in exosomes: Insights into wound healing mechanisms and therapeutic applications. Int J Biol Macromol 2024; 292:139206. [PMID: 39732230 DOI: 10.1016/j.ijbiomac.2024.139206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Revised: 12/16/2024] [Accepted: 12/24/2024] [Indexed: 12/30/2024]
Abstract
Exosomal non-coding RNAs (ncRNAs), including miRNAs, lncRNAs, and circRNAs, have emerged as crucial modulators in cellular signaling, influencing wound healing processes. Stem cell-derived exosomes, which serve as vehicles for these ncRNAs, show remarkable therapeutic potential due to their ability to modulate wound healing stages, from initial inflammation to collagen formation. These ncRNAs act as molecular signals, regulating gene expression and protein synthesis necessary for cellular responses in healing. Wound healing is a complex, staged process involving inflammation, hemostasis, fibroblast proliferation, angiogenesis, and tissue remodeling. Stem cell-derived exosomal ncRNAs enhance these stages by reducing excessive inflammation, promoting anti-inflammatory responses, guiding fibroblast and keratinocyte maturation, enhancing vascularization, and ensuring organized collagen deposition. Their molecular cargo, particularly ncRNAs, specifically targets pathways to aid chronic wound repair and support scarless regeneration. This review delves into the unique composition and signaling roles of Stem cell-derived exosomes and ncRNAs, highlighting their impact across wound healing stages and their potential as innovative therapeutics. Understanding the interaction between exosomal ncRNAs and cellular signaling pathways opens new avenues in regenerative medicine, positioning Stem cell-derived exosomes and their ncRNAs as promising molecular-level interventions in wound healing.
Collapse
Affiliation(s)
- A Gowtham
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Raebareli (NIPER-R), Transit Campus, Bijnor-Sisendi Road, Sarojini Nagar, Near CRPF Base Camp, Lucknow, UP 226002, India
| | - Ravinder K Kaundal
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Raebareli (NIPER-R), Transit Campus, Bijnor-Sisendi Road, Sarojini Nagar, Near CRPF Base Camp, Lucknow, UP 226002, India.
| |
Collapse
|
8
|
Fu E, Pan K, Li Z. Engineering extracellular vesicles for targeted therapeutics in cardiovascular disease. Front Cardiovasc Med 2024; 11:1503830. [PMID: 39749310 PMCID: PMC11693616 DOI: 10.3389/fcvm.2024.1503830] [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: 09/29/2024] [Accepted: 12/09/2024] [Indexed: 01/04/2025] Open
Abstract
Extracellular vesicles (EVs) are nanosized particles secreted by cells that play crucial roles in intercellular communication, especially in the context of cardiovascular diseases (CVDs). These vesicles carry complex cargo, including proteins, lipids, and nucleic acids, that reflects the physiological or pathological state of their cells of origin. Multiomics analysis of cell-derived EVs has provided valuable insights into the molecular mechanisms underlying CVDs by identifying specific proteins and EV-bound targets involved in disease progression. Recent studies have demonstrated that engineered EVs, which are designed to carry specific therapeutic molecules or modified to enhance their targeting capabilities, hold promise for treating CVDs. Analysis of the EV proteome has been instrumental in identifying key proteins that can be targeted or modulated within these engineered vesicles. For example, proteins involved in inflammation, thrombosis, and cardiac remodeling have been identified as potential therapeutic targets. Furthermore, the engineering of EVs to increase their delivery to specific tissues, such as the myocardium, or to modulate their immunogenicity and therapeutic efficacy is an emerging area of research. By leveraging the insights gained from multiomics analyses, researchers are developing EV-based therapies that can selectively target pathological processes in CVDs, offering a novel and potentially more effective treatment strategy. This review integrates the core findings from EV multiomics analysis in the context of CVDs and highlights the potential of engineered EVs in therapeutic applications.
Collapse
Affiliation(s)
- Enze Fu
- School of Medicine, Nankai University, Tianjin, China
- Institute of Ophthalmology, Nankai University, Tianjin, China
| | - Kai Pan
- School of Medicine, Nankai University, Tianjin, China
- Henan Key Laboratory of Cardiac Remodeling and Transplantation, Seventh People's Hospital, Zhengzhou, China
| | - Zongjin Li
- School of Medicine, Nankai University, Tianjin, China
- Institute of Ophthalmology, Nankai University, Tianjin, China
- Henan Key Laboratory of Cardiac Remodeling and Transplantation, Seventh People's Hospital, Zhengzhou, China
- National Key Laboratory of Kidney Diseases, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
9
|
Piccarducci R, Germelli L, Falleni A, Luisotti L, Masciulli B, Signore G, Migone C, Fabiano A, Bizzarri R, Piras AM, Giacomelli C, Marchetti L, Martini C. GFP Farnesylation as a Suitable Strategy for Selectively Tagging Exosomes. ACS APPLIED BIO MATERIALS 2024; 7:8305-8318. [PMID: 39632747 DOI: 10.1021/acsabm.4c01112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2024]
Abstract
Exosomes are small extracellular vesicles (EVs) constituting fully biological, cell-derived nanovesicles with great potential in cell-to-cell communication and drug delivery applications. The current gold standard for EV labeling and tracking is represented by fluorescent lipophilic dyes which, however, importantly lack selectivity, due to their unconditional affinity for lipids. Herein, an alternative EV fluorescent labeling approach is in-depth evaluated, by taking advantage of green fluorescent protein (GFP) farnesylation (GFP-f), a post-translational modification to directly anchor GFP to the EV membrane. The performance of GFP-f is analyzed, in terms of selectivity and efficiency, in several typical EV experimental setups such as delivery in recipient cells, surface engineering, and cargo loading. First, the capability of GFP and GFP-f to label exosomes was compared, showing significantly higher GFP protein levels and fluorescence intensity in GFP-f- than in GFP-labeled exosomes, highlighting the advantage of directly anchoring the GFP to the EV cell membrane. Then, the GFP-f tag was further compared to Vybrant DiD lipophilic dye labeling in exosome uptake studies, by capturing EV intracellular fluorescence in a time- and concentration-dependent manner. The internalization assay revealed a particular ability of GFP-f to monitor the uptake of tagged exosomes into recipient cells, with a significant peak of intensity reached 12 h after administration by GFP-f but not Vybrant-labeled EVs. Finally, the GFP-f labeling capability was challenged in the presence of a surface modification of exosomes and after transfection for siRNA loading. Results showed that both procedures can influence GFP-f performance compared to naïve GFP-f exosomes, although fluorescence is importantly maintained in both cases. Overall, these data provide direct insight into the advantages and limitations of GFP-f as a tagging protein for selectively and accurately tracking the exosome route from isolation to uptake in recipient cells, also in the context of EV bioengineering applications.
Collapse
Affiliation(s)
| | | | - Alessandra Falleni
- Department of Experimental and Clinical Medicine, University of Pisa, 56126 Pisa, Italy
| | | | - Benedetta Masciulli
- Department of Surgical, Medical and Molecular Pathology, and Critical Care Medicine, University of Pisa, 56126 Pisa, Italy
| | - Giovanni Signore
- Department of Biology, University of Pisa, 56126 Pisa, Italy
- Center for Instrument Sharing, University of Pisa, 56126 Pisa, Italy
| | - Chiara Migone
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy
| | - Angela Fabiano
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy
- Center for Instrument Sharing, University of Pisa, 56126 Pisa, Italy
| | - Ranieri Bizzarri
- Department of Surgical, Medical and Molecular Pathology, and Critical Care Medicine, University of Pisa, 56126 Pisa, Italy
- Center for Instrument Sharing, University of Pisa, 56126 Pisa, Italy
- NEST, Istituto Nanoscienze-CNR and Scuola Normale Superiore, 56126 Pisa, Italy
| | - Anna Maria Piras
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy
- Center for Instrument Sharing, University of Pisa, 56126 Pisa, Italy
| | - Chiara Giacomelli
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy
- Center for Instrument Sharing, University of Pisa, 56126 Pisa, Italy
| | - Laura Marchetti
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy
- Center for Instrument Sharing, University of Pisa, 56126 Pisa, Italy
| | - Claudia Martini
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy
- Center for Instrument Sharing, University of Pisa, 56126 Pisa, Italy
| |
Collapse
|
10
|
Sato K, Ogawa K, Tagami T, Ozeki T. Photothermal therapeutic effect by gold nanostars/extracellular vesicles nanocomplex on melanoma cells. J Pharm Sci 2024:S0022-3549(24)00610-5. [PMID: 39694268 DOI: 10.1016/j.xphs.2024.12.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 12/12/2024] [Accepted: 12/12/2024] [Indexed: 12/20/2024]
Abstract
Photothermal therapy (PTT) is a method for treating cancer using the heat generated by light irradiation, often in combination with light-absorbing materials. Efficient PTT requires a drug delivery system to deliver light-absorbing materials to cancerous tissues. Gold nanostars (GNSs) enable efficient PTT through absorbing long-wavelength light. In this study, we established a platform for preparing GNS-loaded extracellular vesicles (EVs), that were expected to provide the combined biological functionality of EVs and photothermal conversion properties. By electrostatically binding cationic polyethylene glycol (PEG)-modified GNSs (PEG-GNSs) to naturally negatively charged EVs, EV/PEG-GNS nanocomplexes (EV-GNSs) were obtained by simply mixing the two components. The prepared EV-GNSs were approximately 200 nm in size and exhibited a negative surface charge. The surface protein marker of EVs, CD63, was detected using western blot, suggesting that the EVs were intact. In vitro evaluation showed that EV-GNSs exhibited better photothermal conversion properties than PEG-GNSs alone. When EV-GNSs were added to the cells, high laser-responsive cytotoxicity was observed following laser irradiation. Thus, EV-GNSs developed in this study may be suitable for PTT using gold-based nanoparticles.
Collapse
Affiliation(s)
- Kazuki Sato
- Drug Delivery and Nano Pharmaceutics, Graduate School of Pharmaceutical Sciences, Nagoya City University, 3-1 Tanabe-dori, Mizuho-ku, Nagoya, Aichi, 467-8603, Japan
| | - Koki Ogawa
- Drug Delivery and Nano Pharmaceutics, Graduate School of Pharmaceutical Sciences, Nagoya City University, 3-1 Tanabe-dori, Mizuho-ku, Nagoya, Aichi, 467-8603, Japan
| | - Tatsuaki Tagami
- Drug Delivery and Nano Pharmaceutics, Graduate School of Pharmaceutical Sciences, Nagoya City University, 3-1 Tanabe-dori, Mizuho-ku, Nagoya, Aichi, 467-8603, Japan
| | - Tetsuya Ozeki
- Drug Delivery and Nano Pharmaceutics, Graduate School of Pharmaceutical Sciences, Nagoya City University, 3-1 Tanabe-dori, Mizuho-ku, Nagoya, Aichi, 467-8603, Japan.
| |
Collapse
|
11
|
Chang X, Wang WX. In vivo bioaccumulation and responses of hemocytes of mussels Perna viridis to microplastics and nanoplastics exposure. JOURNAL OF HAZARDOUS MATERIALS 2024; 480:135939. [PMID: 39321482 DOI: 10.1016/j.jhazmat.2024.135939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 09/09/2024] [Accepted: 09/21/2024] [Indexed: 09/27/2024]
Abstract
Growing micro- and nano-plastic (MNPs) pollution in the environment poses a threat to marine animals. Due to their excellent filtration capacity, bivalves can easily ingest MNPs, which could be translocated to open circulation system with potential risks. In the present study, the accumulation and elimination of MNPs (200 nm and 1 µm) in the mussel hemolymph serum and hemocytes were firstly quantified, and the differential sensitiveresponses of two subpopulations of hemocytes were then explored by in vivo exposure under environmentally relevant concentration of MNPs (200 µg/L). We demonstrated that MNPs were readily translocated into hemolymph serum, but were immediately followed by efficient internalization by hemocytes. Remarkably, concentrations of MNPs in hemolymph were only 0.63 and 0.39 times lower than the ambient exposure concentration. Granulocytes displayed a much higher potential of accumulating MNPs than the agranulocytes. MPs were more readily internalized by granulocytes, with their estimated maximum bioaccumulation factor (BCF) of 0.29 L/g. Due to the primary function of phagocytic encapsulation of MNPs by granulocytes, lysosome features especially the decline of subsequent lysosome membrane potential could be a potential sensitive biomarker in response to MNPs exposure. Our results provided insights on the bioaccumulation of MNPs at the cellular levels in marine bivalves.
Collapse
Affiliation(s)
- Xinyi Chang
- School of Energy and Environment and State Key Laboratory of Marine Pollution, City University of Hong Kong, Kowloon, Hong Kong; Research Centre for the Oceans and Human Health, City University of Hong Kong Shenzhen Research Institute, Shenzhen 518057, China
| | - Wen-Xiong Wang
- School of Energy and Environment and State Key Laboratory of Marine Pollution, City University of Hong Kong, Kowloon, Hong Kong; Research Centre for the Oceans and Human Health, City University of Hong Kong Shenzhen Research Institute, Shenzhen 518057, China.
| |
Collapse
|
12
|
Dong G, Douanne N, Fernandez-Prada C, Olivier M. Unique Leishmania mexicana clones secrete populations of extracellular vesicles with unique protein profile and variable infectious capability. Front Cell Infect Microbiol 2024; 14:1443262. [PMID: 39703372 PMCID: PMC11655471 DOI: 10.3389/fcimb.2024.1443262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 11/12/2024] [Indexed: 12/21/2024] Open
Abstract
The study of extracellular vesicles has become an incredibly important field of study, but the inherent heterogeneity of these vesicles continues to make their study challenging. The genetic variability and well-documented protocols for the growth and vesicle isolation from Leishmania parasites provide a unique opportunity to compare the heterogeneity of different populations secreted by Leishmania clones. Leishmania mexicana was cultured on solid SDM agar plates and 8 clonal colonies were selected. The EVs collected from the liquid cultures of these 8 clones were assessed by NTA, TEM, and proteomic analysis. We found that all 8 clonal L. mexicana cultures were visually indistinguishable from each other and had similar growth rate, and these physical similarities extended to their EVs. However, proteomic analysis reveals that the EVs collected have unique protein profiles compared to each other and EVs isolated from a heterogeneous liquid culture of L. mexicana. We selected 3 clonal EVs for further mouse infection experiments and found that EVs from CL7 L. mexicana consistently caused reduced footpad swelling in C57BL6 mice footpads compared to EVs from CL1, CL8, and heterogenous L. mexicana. This trend was not observed when infecting Balb/C mice and C57BL6 with the parasites alone, with only CL1 L. mexicana causing significantly increased infection in Balb/c mice. Our results together show that EVs isolated from different clonal colonies of L. mexicana have distinct differences in protein cargo which can lead to varying outcomes on Leishmania infection. Further evaluation will be needed to determine the underlying mechanisms behind this and verify that differences observed in infectivity are directly caused by variations between our L. mexicana clones, especially genetic sequencing and immunoblotting to validate our results.
Collapse
Affiliation(s)
- George Dong
- Infectious Diseases and Immunity in Global Health Program, Research Institute of McGill University Health Centre, Montréal, QC, Canada
| | - Noélie Douanne
- Department of Pathology and Microbiology, Faculty of Veterinary Medicine, Université de Montréal, Saint-Hyacinthe, QC, Canada
| | - Christopher Fernandez-Prada
- Department of Pathology and Microbiology, Faculty of Veterinary Medicine, Université de Montréal, Saint-Hyacinthe, QC, Canada
| | - Martin Olivier
- Infectious Diseases and Immunity in Global Health Program, Research Institute of McGill University Health Centre, Montréal, QC, Canada
- Department of Microbiology and Immunology, McGill University, Montréal, QC, Canada
| |
Collapse
|
13
|
Jung H, Jung Y, Seo J, Bae Y, Kim HS, Jeong W. Roles of extracellular vesicles from mesenchymal stem cells in regeneration. Mol Cells 2024; 47:100151. [PMID: 39547584 DOI: 10.1016/j.mocell.2024.100151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 11/09/2024] [Accepted: 11/10/2024] [Indexed: 11/17/2024] Open
Abstract
Mesenchymal stem cells (MSCs) are highly valued in regenerative medicine due to their ability to self-renew and differentiate into various cell types. Their therapeutic benefits are primarily due to their paracrine effects, in particular through extracellular vesicles (EVs), which are related to intercellular communication. Recent advances in EV production and extraction technologies highlight the potential of MSC-derived EVs (MSC-EVs) in tissue engineering and regenerative medicine. MSC-EVs offer several advantages over traditional cell therapies, including reduced toxicity and immunogenicity compared with whole MSCs. EVs carrying functional molecules such as growth factors, cytokines, and miRNAs play beneficial roles in tissue repair, fibrosis treatment, and scar prevention by promoting angiogenesis, skin cell migration, proliferation, extracellular matrix remodeling, and reducing inflammation. Despite the potential of MSC-EVs, there are several limitations to their use, including variability in quality, the need for standardized methods, low yield, and concerns about the composition of EVs and the potential risks. Overall, MSC-EVs are a promising alternative to cell-based therapies, and ongoing studies aim to understand their actions and optimize their use for better clinical outcomes in wound healing and skin regeneration.
Collapse
Affiliation(s)
- Hyeseong Jung
- Department of Biomedical Science, Catholic Kwandong University, Gangneung 25601, Republic of Korea
| | - Yuyeon Jung
- Department of Dental Hygiene, Catholic Kwandong University, Gangneung 25601, Republic of Korea
| | - Junsik Seo
- Department of Biomedical Science, Catholic Kwandong University, Gangneung 25601, Republic of Korea
| | - Yeongju Bae
- Department of Biomedical Science, Catholic Kwandong University, Gangneung 25601, Republic of Korea; Research Center for Marine Bio-Food and Medicine, Catholic Kwandong University, Gangneung 25601, Republic of Korea
| | - Han-Soo Kim
- Department of Biomedical Science, Catholic Kwandong University, Gangneung 25601, Republic of Korea
| | - Wooyoung Jeong
- Department of Biomedical Science, Catholic Kwandong University, Gangneung 25601, Republic of Korea; Research Center for Marine Bio-Food and Medicine, Catholic Kwandong University, Gangneung 25601, Republic of Korea.
| |
Collapse
|
14
|
Oh C, Mazan-Mamczarz K, Gorospe M, Noh JH, Kim KM. Impact of UPF2 on the levels of CD81 on extracellular vesicles. Front Cell Dev Biol 2024; 12:1469080. [PMID: 39655046 PMCID: PMC11625909 DOI: 10.3389/fcell.2024.1469080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 10/29/2024] [Indexed: 12/12/2024] Open
Abstract
Extracellular vesicles (EVs) are involved in cell-to-cell communication. Following uptake, EV cargo molecules, including DNA, RNA, lipids, and proteins, influence gene expression and molecular signaling in recipient cells. Although various studies have identified disease-specific EV molecules, further research into their biogenesis and secretion mechanisms is needed for clinical application. Here, we investigated the role of UPF2 in regulating the biogenesis and components of EVs. Notably, UPF2 promoted the expression of CD81, a membrane protein marker of EVs, as UPF2 silencing decreased CD81 levels in EVs, both inside the cell and secreted. In contrast, the expression levels of CD63 increased, without altering the size or numbers of EVs. In addition, reducing UPF2 levels did not affect the total number of EVs but lowered production of CD81-positive EVs and reduced the efficiency of uptake by recipient cells. Collectively, our findings uncover a novel function for UPF2 in regulating the production of CD81 and changing EV properties.
Collapse
Affiliation(s)
- Chaehwan Oh
- Department of Biological Sciences, Chungnam National University, Daejeon, Republic of Korea
| | - Krystyna Mazan-Mamczarz
- Laboratory of Genetics and Genomics, National Institute on Aging Intramural Research Program, National Institutes of Health, Baltimore, MD, United States
| | - Myriam Gorospe
- Laboratory of Genetics and Genomics, National Institute on Aging Intramural Research Program, National Institutes of Health, Baltimore, MD, United States
| | - Ji Heon Noh
- Molecular Aging Biology Laboratory (MABL), Department of Biochemistry, College of Natural Science, Chungnam National University, Daejeon, Republic of Korea
| | - Kyoung Mi Kim
- Department of Biological Sciences, Chungnam National University, Daejeon, Republic of Korea
| |
Collapse
|
15
|
Noureddine N, Holtzhauer G, Wawrzyniak P, Srikanthan P, Krämer SD, Rogler G, Lucchinetti E, Zaugg M, Hersberger M. Size of lipid emulsion droplets influences metabolism in human CD4 + T cells. Biochem Biophys Res Commun 2024; 733:150680. [PMID: 39278094 DOI: 10.1016/j.bbrc.2024.150680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 09/03/2024] [Accepted: 09/09/2024] [Indexed: 09/17/2024]
Abstract
SCOPE Triglyceride-based lipid emulsions are critical for total parenteral nutrition (TPN), but their long-term use has adverse effects, such as severe liver dysfunction necessitating improved formulations. This study compares the uptake mechanism and intracellular fate of novel glycerol-stabilized nano-sized lipid emulsions with conventional emulsions in CD4+ T cells, focusing on their impact on cellular metabolism. METHODS AND RESULTS Nanoemulsions were formulated with increased glycerol content. Uptake of emulsions in primary human CD4+ T cells was investigated using different endocytic blockers, then quantified by flow cytometry, and visualized by confocal microscopy. To investigate emulsion intracellular fate, fatty acids in membrane phospholipids were quantified by GC-MS/MS and cellular metabolism was assessed by Seahorse technology. Results show T cells internalize both conventional and nano-sized emulsions using macropinocytosis. Fatty acids from emulsions are stored as neutral lipids in intracellular vesicles and are incorporated into phospholipids of cellular membranes. However, only nanoemulsions additionally use clathrin-mediated endocytosis and deliver fatty acids to mitochondria for increased β-oxidation. CONCLUSIONS Size of lipid emulsion droplets significantly influences their uptake and subsequent metabolism in CD4+ T cells. Our results highlight the potential for improved nutrient utilization with nanoemulsions in TPN formulations possibly leading to less adverse effects.
Collapse
Affiliation(s)
- Nazek Noureddine
- Division of Clinical Chemistry and Biochemistry, Children's Research Center, University Children's Hospital Zurich and University of Zurich, Zurich, Switzerland; Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland.
| | - Gregory Holtzhauer
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, Zurich, Switzerland
| | - Paulina Wawrzyniak
- Department of Gastroenterology and Hepatology, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Pakeerathan Srikanthan
- Division of Clinical Chemistry and Biochemistry, Children's Research Center, University Children's Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Stefanie D Krämer
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, Zurich, Switzerland
| | - Gerhard Rogler
- Department of Gastroenterology and Hepatology, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Eliana Lucchinetti
- Department of Anesthesiology and Pain Medicine and Cardiovascular Research Centre, University of Alberta, Edmonton, Canada
| | - Michael Zaugg
- Department of Anesthesiology and Pain Medicine and Cardiovascular Research Centre, University of Alberta, Edmonton, Canada; Department of Pharmacology, University of Alberta, Edmonton, Canada
| | - Martin Hersberger
- Division of Clinical Chemistry and Biochemistry, Children's Research Center, University Children's Hospital Zurich and University of Zurich, Zurich, Switzerland; Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
16
|
Fang L, Gu W, Li R, Chen C, Cai S, Luozhong S, Chen M, Hsu A, Tsai YC, Londhe K, Jiang S. Controlling Circular RNA Encapsulation within Extracellular Vesicles for Gene Editing and Protein Replacement. ACS NANO 2024; 18:30378-30387. [PMID: 39445782 DOI: 10.1021/acsnano.4c07530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2024]
Abstract
Extracellular vesicles (EVs) are a population of vesicular bodies originating from cells, and EVs have been proven to have the potential to deliver different cargos, such as RNAs. However, conventional methods are not able to encapsulate long RNAs into EVs efficiently or may compromise the integrity of EVs. In this study, we have devised a strategy to encapsulate long circRNAs (>1000 nt) into EVs by harnessing the sorting mechanisms of cells. This strategy utilizes the inherent richness of circular RNAs in EVs and a genetic engineering method to increase the cytoplasmic concentration of target circRNAs, facilitating highly efficient RNA back-splicing to drive the circularization of RNAs. This allows target circRNAs to load into EVs with high efficiency. Furthermore, we demonstrate the practical applications of this strategy, showing that these circRNAs can be delivered by EVs to recipient cells for protein expression and to mice for gene editing.
Collapse
Affiliation(s)
- Liang Fang
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York 14853, United States
| | - Wenchao Gu
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York 14853, United States
| | - Ruoxin Li
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York 14853, United States
| | - Chaoxin Chen
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York 14853, United States
| | - Simian Cai
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York 14853, United States
| | - Sijin Luozhong
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York 14853, United States
| | - Michelle Chen
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York 14853, United States
| | - Annie Hsu
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York 14853, United States
| | - Yi-Chih Tsai
- Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, New York 14853, United States
| | - Ketaki Londhe
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York 14853, United States
| | - Shaoyi Jiang
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York 14853, United States
| |
Collapse
|
17
|
Ma M, Li X, Jing M, Zhang P, Zhang M, Wang L, Liang X, Jiang Y, Li J, He J, Wang X, Lin M, Wang L, Fan J. Enhanced Tumor-Targeted Delivery of Arginine-Rich Peptides via a Positive Feedback Loop Orchestrated by Piezo1/integrin β1 Signaling Axis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2409081. [PMID: 39258781 PMCID: PMC11558097 DOI: 10.1002/advs.202409081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Indexed: 09/12/2024]
Abstract
Peptide-based drugs hold great potential for cancer treatment, and their effectiveness is driven by mechanisms on how peptides target cancer cells and escape from potential lysosomal entrapment post-endocytosis. Yet, the mechanisms remain elusive, which hinder the design of peptide-based drugs. Here hendeca-arginine peptides (R11) are synthesized for targeted delivery in bladder carcinoma (BC), investigated the targeting efficiency and elucidated the mechanism of peptide-based delivery, with the aim of refining the design and efficacy of peptide-based therapeutics. It is demonstrated that the over-activated Piezo1/integrin β1 (ITGB1) signaling axis significantly facilitates tumor-targeted delivery of R11 peptides via macropinocytosis. Furthermore, R11 peptides formed hydrogen bonds with integrin β1, facilitating targeting and penetration into tumor cells. Additionally, R11 peptides protected integrin β1 from lysosome degradation, promoting its recycling from cytoplasm to membrane. Moreover, this findings establish a positive feedback loop wherein R11 peptides activate Piezo1 by increasing membrane fusion, promoting Ca2+ releasing and resulting in enhanced integrin β1-mediated endocytosis in both orthotopic models and clinical tissues, demonstrating effective tumor-targeted delivery. Eventually, the Piezo1/integrin β1 signaling axis promoted cellular uptake and transport of peptides, establishing a positive feedback loop, promoting mechanical delivery to cancer and offering possibilities for drug modification in cancer therapy.
Collapse
Affiliation(s)
- Minghai Ma
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Department of Urology, The First Affiliated HospitalXi'an Jiaotong UniversityXi'an710061China
| | - Xing Li
- Department of Thoracic Surgery, Tangdu HospitalAir Force Medical UniversityXi'an710038China
| | - Minxuan Jing
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Department of Urology, The First Affiliated HospitalXi'an Jiaotong UniversityXi'an710061China
| | - Pu Zhang
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Department of Urology, The First Affiliated HospitalXi'an Jiaotong UniversityXi'an710061China
| | - Mengzhao Zhang
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Department of Urology, The First Affiliated HospitalXi'an Jiaotong UniversityXi'an710061China
| | - Lu Wang
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Department of Urology, The First Affiliated HospitalXi'an Jiaotong UniversityXi'an710061China
| | - Xiao Liang
- Department of Thoracic Surgery, Tangdu HospitalAir Force Medical UniversityXi'an710038China
| | - Yunzhong Jiang
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Department of Urology, The First Affiliated HospitalXi'an Jiaotong UniversityXi'an710061China
| | - Jianpeng Li
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Department of Urology, The First Affiliated HospitalXi'an Jiaotong UniversityXi'an710061China
| | - Jiale He
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Department of Urology, The First Affiliated HospitalXi'an Jiaotong UniversityXi'an710061China
| | - Xinyang Wang
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Department of Urology, The First Affiliated HospitalXi'an Jiaotong UniversityXi'an710061China
| | - Min Lin
- Key Laboratory of Biomedical Information Engineering, Ministry of Education, Bioinspired Engineering and Biomechanics Center (BEBC), School of Life Science and TechnologyXi'an Jiaotong UniversityXi'an710049China
| | - Lei Wang
- Department of Thoracic Surgery, Tangdu HospitalAir Force Medical UniversityXi'an710038China
| | - Jinhai Fan
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Department of Urology, The First Affiliated HospitalXi'an Jiaotong UniversityXi'an710061China
| |
Collapse
|
18
|
Pinheiro SKDP, Pontes MDS, Miguel TBAR, Grillo R, Souza Filho AGD, Miguel EDC. Nanoparticles and plants: A focus on analytical characterization techniques. PLANT SCIENCE : AN INTERNATIONAL JOURNAL OF EXPERIMENTAL PLANT BIOLOGY 2024; 348:112225. [PMID: 39142607 DOI: 10.1016/j.plantsci.2024.112225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 07/05/2024] [Accepted: 08/11/2024] [Indexed: 08/16/2024]
Abstract
Nanotechnology has brought about significant progress through the use of goods based on nanomaterials. However, concerns remain about the accumulation of these materials in the environment and their potential toxicity to living organisms. Plants have the ability to take in nanomaterials (NMs), which can cause changes in their physiology and morphology. On the other hand, nanoparticles (NPs) have been used to increase plant development and control pests in agriculture by including them into agrochemicals. The challenges of the interaction, internalization, and accumulation of NMs within plant tissues are enormous, mainly because of the various characteristics of NMs and the absence of reliable analytical tools. As our knowledge of the interactions between NMs and plant cells expands, we are able to create novel NMs that are tailored, targeted, and designed to be safe, thus minimizing the environmental consequences of nanomaterials. This review provides a thorough examination and comparison of the main microscopy techniques, spectroscopic methods, and far-field super-resolution methodologies used to examine nanomaterials within the cell walls of plants.
Collapse
Affiliation(s)
- Sergimar Kennedy de Paiva Pinheiro
- Biomaterials Laboratory, Department of Metallurgical Engineering and Materials and Analytical Center, Federal University of Ceará (UFC), Fortaleza, CE, Brazil
| | - Montcharles da Silva Pontes
- Optics and Photonics Group, SISFOTON Lab, Institute of Physics, Federal University of Mato Grosso do Sul (UFMS), Campo Grande, MS, Brazil
| | | | - Renato Grillo
- Environmental Nanochemistry Group, Department of Physics and Chemistry, São Paulo State University (UNESP), Ilha Solteira, SP, Brazil
| | | | - Emilio de Castro Miguel
- Biomaterials Laboratory, Department of Metallurgical Engineering and Materials and Analytical Center, Federal University of Ceará (UFC), Fortaleza, CE, Brazil.
| |
Collapse
|
19
|
Ansa‐Addo EA, Pathak P, McCrossan MV, Volpato Rossi I, Abdullahi M, Stratton D, Lange S, Ramirez MI, Inal JM. Monocyte-derived extracellular vesicles, stimulated by Trypanosoma cruzi, enhance cellular invasion in vitro via activated TGF-β1. J Extracell Vesicles 2024; 13:e70014. [PMID: 39611395 PMCID: PMC11605483 DOI: 10.1002/jev2.70014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 09/06/2024] [Accepted: 10/24/2024] [Indexed: 11/30/2024] Open
Abstract
During cell invasion, large Extracellular Vesicle (lEV) release from host cells was dose-dependently triggered by Trypanosoma cruzi metacyclic trypomastigotes (Mtr). This lEV release was inhibited when IP3-mediated Ca2+ exit from the ER and further Ca2+ entry from plasma membrane channels was blocked, but whilst any store-independent Ca2+ entry (SICE) could continue unabated. That lEV release was equally inhibited if all entry from external sources was blocked by chelation of external Ca2+ points to the major contributor to Mtr-triggered host cell lEV release being IP3/store-mediated Ca2+ release, SICE playing a minor role. Host cell lEVs were released through Mtr interaction with host cell lipid raft domains, integrins, and mechanosensitive ion channels, whereupon [Ca2+]cyt increased (50 to 750 nM) within 15 s. lEV release and cell entry of T. cruzi, which increased up to 30 and 60 mpi, respectively, as well as raised actin depolymerization at 60 mpi, were all reduced by TRPC inhibitor, GsMTx-4. Vesicle release and infection was also reduced with RGD peptide, methyl-β-cyclodextrin, knockdown of calpain and with the calpain inhibitor, calpeptin. Restoration of lEV levels, whether with lEVs from infected or uninfected epithelial cells, did not restore invasion, but supplementation with lEVs from infected monocytes, did. We provide evidence of THP-1 monocyte-derived lEV interaction with Mtr (lipid mixing by R18-dequenching; flow cytometry showing transfer to Mtr of R18 from R18-lEVs and of LAP(TGF-β1). Active, mature TGF-β1 (at 175 pg/×105 in THP-1 lEVs) was detected in concentrated lEV-/cell-free supernatant by western blotting, only after THP-1 lEVs had interacted with Mtr. The TGF-β1 receptor (TβRI) inhibitor, SB-431542, reduced the enhanced cellular invasion due to monocyte-lEVs.
Collapse
Affiliation(s)
- Ephraim A. Ansa‐Addo
- School of Human Sciences, Cell Communication in Disease PathologyLondon Metropolitan UniversityLondonUK
- Pelotonia Institute for Immuno‐Oncology, Department of Internal MedicineThe Ohio State University Comprehensive Cancer CenterColumbusOhioUSA
| | - Paras Pathak
- School of Human Sciences, Cell Communication in Disease PathologyLondon Metropolitan UniversityLondonUK
- Medical Research Council HarwellHarwell Science and Innovation Campus, Genotyping CoreOxfordshireUK
| | | | - Izadora Volpato Rossi
- School of Human Sciences, Cell Communication in Disease PathologyLondon Metropolitan UniversityLondonUK
- School of Life and Medical Sciences, Biosciences Research GroupUniversity of HertfordshireHatfieldUK
- Carlos Chagas InstituteFundacao Oswaldo Cruz, (FIOCRUZ‐PR)CuritibaBrazil
- Postgraduate Program in Cellular and Molecular BiologyFederal University of ParanáCuritibaBrazil
| | - Mahamed Abdullahi
- School of Human Sciences, Cell Communication in Disease PathologyLondon Metropolitan UniversityLondonUK
- National Mycobacterium Reference Service‐South (NMRS‐South) ColindaleLondonUK
| | - Dan Stratton
- School of Life, Health & Chemical SciencesThe Open UniversityMilton KeynesUK
| | - Sigrun Lange
- Tissue Architecture and Regeneration Research Group, School of Life SciencesUniversity of WestminsterLondonUK
- University College London, Institute of Women's HealthLondonUK
| | - Marcel I. Ramirez
- Carlos Chagas InstituteFundacao Oswaldo Cruz, (FIOCRUZ‐PR)CuritibaBrazil
| | - Jameel M. Inal
- School of Human Sciences, Cell Communication in Disease PathologyLondon Metropolitan UniversityLondonUK
- School of Life and Medical Sciences, Biosciences Research GroupUniversity of HertfordshireHatfieldUK
| |
Collapse
|
20
|
Jackson Cullison SR, Flemming JP, Karagoz K, Wermuth PJ, Mahoney MG. Mechanisms of extracellular vesicle uptake and implications for the design of cancer therapeutics. JOURNAL OF EXTRACELLULAR BIOLOGY 2024; 3:e70017. [PMID: 39483807 PMCID: PMC11522837 DOI: 10.1002/jex2.70017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 09/11/2024] [Accepted: 10/14/2024] [Indexed: 11/03/2024]
Abstract
The translation of pre-clinical anti-cancer therapies to regulatory approval has been promising, but slower than hoped. While innovative and effective treatments continue to achieve or seek approval, setbacks are often attributed to a lack of efficacy, failure to achieve clinical endpoints, and dose-limiting toxicities. Successful efforts have been characterized by the development of therapeutics designed to specifically deliver optimal and effective dosing to tumour cells while minimizing off-target toxicity. Much effort has been devoted to the rational design and application of synthetic nanoparticles to serve as targeted therapeutic delivery vehicles. Several challenges to the successful application of this modality as delivery vehicles include the induction of a protracted immune response that results in their rapid systemic clearance, manufacturing cost, lack of stability, and their biocompatibility. Extracellular vesicles (EVs) are a heterogeneous class of endogenous biologically produced lipid bilayer nanoparticles that mediate intercellular communication by carrying bioactive macromolecules capable of modifying cellular phenotypes to local and distant cells. By genetic, chemical, or metabolic methods, extracellular vesicles (EVs) can be engineered to display targeting moieties on their surface while transporting specific cargo to modulate pathological processes following uptake by target cell populations. This review will survey the types of EVs, their composition and cargoes, strategies employed to increase their targeting, uptake, and cargo release, and their potential as targeted anti-cancer therapeutic delivery vehicles.
Collapse
Affiliation(s)
| | - Joseph P. Flemming
- Rowan‐Virtua School of Osteopathic MedicineRowan UniversityStratfordNew JerseyUSA
| | - Kubra Karagoz
- Departments of PharmacologyPhysiology, and Cancer Biology, Thomas Jefferson UniversityPhiladelphiaPennsylvaniaUSA
| | | | - Mỹ G. Mahoney
- Departments of PharmacologyPhysiology, and Cancer Biology, Thomas Jefferson UniversityPhiladelphiaPennsylvaniaUSA
- Department of Otolaryngology – Head and Neck SurgeryThomas Jefferson UniversityPhiladelphiaPennsylvaniaUSA
| |
Collapse
|
21
|
Jhaveri JR, Khare P, Paul Pinky P, Kamte YS, Chandwani MN, Milosevic J, Abraham N, Sun M, Stolz DB, Dave KM, Zheng SY, O'Donnell L, Manickam DS. Low pinocytic brain endothelial cells primarily utilize membrane fusion to internalize extracellular vesicles. Eur J Pharm Biopharm 2024; 204:114500. [PMID: 39303949 DOI: 10.1016/j.ejpb.2024.114500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 08/30/2024] [Accepted: 09/10/2024] [Indexed: 09/22/2024]
Abstract
Extracellular vesicles (EVs) are an emerging class of drug carriers and are primarily reported to be internalized into recipient cells via a combination of endocytic routes such as clathrin-mediated, caveolae-mediated and macropinocytosis pathways. In this work, (1) we investigated potential effects of homotypic vs. heterotypic interactions by studying the cellular uptake of homologous EVs (EV donor cells and recipient cells of the same type) vs. heterologous EVs (EV donor cells and recipient cells of different types) and (2) determined the route of EV internalization into low pinocytic/hard-to-deliver cell models such as brain endothelial cells (BECs). Homotypic interactions led to a greater extent of uptake into the recipient BECs compared to heterotypic interactions. However, we did not see a complete reduction in EV uptake into recipient BECs when endocytic pathways were blocked using pharmacological inhibitors and our findings from a R18-based fusion assay suggest that EVs primarily use membrane fusion to enter low-pinocytic recipient BECs instead of relying on endocytosis. Lipophilic PKH67 dye-labeled EVs but not intravesicular esterase-activated calcein ester-labeled EVs severely reduced particle uptake into BECs while phagocytic macrophages internalized EVs labeled with both dyes to comparable extents. Our results also highlight the importance of carefully choosing labeling dye chemistry to study EV uptake, especially in the case of low pinocytic cells such as BECs.
Collapse
Affiliation(s)
- Jhanvi R Jhaveri
- Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA, United States
| | - Purva Khare
- Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA, United States
| | - Paromita Paul Pinky
- Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA, United States
| | - Yashika S Kamte
- Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA, United States
| | - Manisha N Chandwani
- Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA, United States
| | - Jadranka Milosevic
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA, United States; Captis Diagnostics Inc., Pittsburgh, PA, United States
| | - Nevil Abraham
- Unified Flow Core, University of Pittsburgh, Pittsburgh, PA, United States
| | - Ming Sun
- Center for Biologic Imaging, University of Pittsburgh Medical School, Pittsburgh, PA, United States
| | - Donna B Stolz
- Center for Biologic Imaging, University of Pittsburgh Medical School, Pittsburgh, PA, United States
| | - Kandarp M Dave
- Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA, United States
| | - Si-Yang Zheng
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA, United States
| | - Lauren O'Donnell
- Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA, United States
| | - Devika S Manickam
- Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA, United States.
| |
Collapse
|
22
|
Wang X, Zhang Z, Qi Y, Zhang Z, Zhang Y, Meng K, Yuan J, Quan F. Study of the uptake mechanism of two small extracellular vesicle subtypes by granulosa cells. Anim Reprod Sci 2024; 270:107576. [PMID: 39178587 DOI: 10.1016/j.anireprosci.2024.107576] [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: 04/11/2024] [Revised: 07/25/2024] [Accepted: 08/10/2024] [Indexed: 08/26/2024]
Abstract
As a new mechanism of intercellular communication, the uptake of extracellular vesicles (EVs) by receptor cells has become a hot topic in the field. Previously, research on the uptake of EVs has focused on the mechanism of small EVs (sEVs, also known as exosomes). As sEVs represent a mixed heterogeneous population, the issue of whether there are different uptake mechanisms for different subsets of sEVs by recipient cells urgently need to be addressed. There are EVs in follicular fluid, which play an important role in the communication between follicular cells and the development of oocytes. Previously, we isolated two subtypes of sEVs in follicular fluid: low density-sEVs (LD-sEVs) and high density-sEVs (HD-sEVs). The current study aimed to explore the uptake characteristics of these two subtypes of sEVs by granulosa cells. First, PKH67 was used to label the two sEVs subtypes, and we observed their uptake by granulosa cells using confocal microscopy and flow cytometry. We then explored the specific mechanisms underlying uptake of these two sEV subtypes by granulosa cells using specific inhibitors and RNA interference. The results showed that granulosa cells took up both kinds of sEVs through a clathrin-independent pathway. In addition to requiring caveolin, cholesterol, and Na+/H+ exchange, the uptake of HD-sEVs also depended on the activity of tyrosine kinase and phosphoinositide 3-kinase. A better understanding of the mechanism of granulosa cell uptake of different subtypes of sEVs in follicular fluid is of considerable significance leading to more accurate use of EVs for targeted treatment of infertility and other related diseases.
Collapse
Affiliation(s)
- Xiaomei Wang
- College of Basic Medicine, Jining Medical University, Jining 272000, China; College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Zihan Zhang
- College of Second Clinical Medical, Jining Medical University, Jining 272000, China
| | - Yuanmin Qi
- College of Clinical Medicine, Jining Medical University, Jining 272000, China
| | - Zhimin Zhang
- College of Clinical Medicine, Jining Medical University, Jining 272000, China
| | - Yixin Zhang
- College of Second Clinical Medical, Jining Medical University, Jining 272000, China
| | - Kai Meng
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China; Lin He's Academician Workstation of New Medicine and Clinical Translation, Jining Medical University, Jining 272000, China
| | - Jinxiang Yuan
- Lin He's Academician Workstation of New Medicine and Clinical Translation, Jining Medical University, Jining 272000, China.
| | - Fusheng Quan
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China.
| |
Collapse
|
23
|
Sabatke B, Rossi IV, Sana A, Bonato LB, Ramirez MI. Extracellular vesicles biogenesis and uptake concepts: A comprehensive guide to studying host-pathogen communication. Mol Microbiol 2024; 122:613-629. [PMID: 37758682 DOI: 10.1111/mmi.15168] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 08/30/2023] [Accepted: 09/04/2023] [Indexed: 09/29/2023]
Abstract
The study of host-pathogen interactions has increased considerably in recent decades. This intercellular communication has been mediated by extracellular vesicles (EVs) that play an important role during the interaction. EVs are particles of lipid bilayer and described in different types of cells, eukaryotic or prokaryotic. Depending on their biogenesis they are described as exosomes (derived from multivesicular bodies) and microvesicles (derived from the plasma membrane). The EVs carry biomolecules, including nucleic acids, lipids, and proteins that can be released or internalized by other cells in different pathways (endocytosis, macropinocytosis, phagocytosis, or membrane fusion) in the process described as uptake. The balance between biogenesis and uptake of EVs could modify physiological and pathophysiological processes of the cell. This review is focusing on the dynamic roles of release and capture of EVs during host-pathogen interaction. We also do a critical analysis of methodologies for obtaining and analyzing EVs. Finally, we draw attention to critical points to be considered in EV biogenesis and uptake studies.
Collapse
Affiliation(s)
- Bruna Sabatke
- Graduate Program in Microbiology, Pathology and Parasitology, Federal University of Paraná, Curitiba, Brazil
- EVAHPI - Extracellular Vesicles and Host-Parasite Interactions Research Group, Carlos Chagas Institute (Fiocruz-PR), Curitiba, Brazil
| | - Izadora Volpato Rossi
- EVAHPI - Extracellular Vesicles and Host-Parasite Interactions Research Group, Carlos Chagas Institute (Fiocruz-PR), Curitiba, Brazil
- Graduate Program in Cell and Molecular Biology, Federal University of Paraná, Curitiba, Brazil
| | - Abel Sana
- EVAHPI - Extracellular Vesicles and Host-Parasite Interactions Research Group, Carlos Chagas Institute (Fiocruz-PR), Curitiba, Brazil
- Graduate Program in Cell and Molecular Biology, Federal University of Paraná, Curitiba, Brazil
| | - Leticia Bassani Bonato
- Graduate Program in Microbiology, Pathology and Parasitology, Federal University of Paraná, Curitiba, Brazil
- EVAHPI - Extracellular Vesicles and Host-Parasite Interactions Research Group, Carlos Chagas Institute (Fiocruz-PR), Curitiba, Brazil
| | - Marcel I Ramirez
- EVAHPI - Extracellular Vesicles and Host-Parasite Interactions Research Group, Carlos Chagas Institute (Fiocruz-PR), Curitiba, Brazil
| |
Collapse
|
24
|
Zhang Y, Lan M, Chen Y. Minimal Information for Studies of Extracellular Vesicles (MISEV): Ten-Year Evolution (2014-2023). Pharmaceutics 2024; 16:1394. [PMID: 39598518 PMCID: PMC11597804 DOI: 10.3390/pharmaceutics16111394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 10/25/2024] [Accepted: 10/27/2024] [Indexed: 11/29/2024] Open
Abstract
In the tenth year since the first edition of MISEV was released in 2014, MISEV2023 has been reported in 2024 with the aim of refining the standard and improving the rigor, reproducibility, and transparency of extracellular vesicle (EV) research to clarify the requirements for experimental design of EVs, emphasize the importance of reproducible experimental results as well as encouraging openness of experimental information. The release of MISEV has significantly contributed to the quality of research in the field of EVs, which creates a more reliable research environment. However, despite the important role of MISEV, there is still a need for the EV researchers to continue to push for the widespread implementation of the guidelines to meet the evolving nature and challenges of EV research. The evolution of EV research and the attention it receives have grown exponentially over time, as has the number of people involved in the writing of MISEV. Here, this review briefly summarizes the evolution of the three editions of MISEV, aiming to recall MISEV2014 and MISEV2018 while learning about the latest release, MISEV2023, to gain a deeper understanding of the content, and to provide a quick note for beginners who want to learn about MISEV and explore the EV world.
Collapse
Affiliation(s)
- Yuan Zhang
- School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang 330006, China; (Y.Z.); (M.L.)
- Jiangxi Key Laboratory for Microscale Interdisciplinary Study, Institute for Advanced Study, Nanchang University, Nanchang 330031, China
| | - Mengyi Lan
- School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang 330006, China; (Y.Z.); (M.L.)
- Jiangxi Key Laboratory for Microscale Interdisciplinary Study, Institute for Advanced Study, Nanchang University, Nanchang 330031, China
| | - Yong Chen
- School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang 330006, China; (Y.Z.); (M.L.)
- Jiangxi Key Laboratory for Microscale Interdisciplinary Study, Institute for Advanced Study, Nanchang University, Nanchang 330031, China
| |
Collapse
|
25
|
Bader J, Brigger F, Leroux JC. Extracellular vesicles versus lipid nanoparticles for the delivery of nucleic acids. Adv Drug Deliv Rev 2024; 215:115461. [PMID: 39490384 DOI: 10.1016/j.addr.2024.115461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 10/21/2024] [Accepted: 10/23/2024] [Indexed: 11/05/2024]
Abstract
Extracellular vesicles (EVs) are increasingly investigated for delivering nucleic acid (NA) therapeutics, leveraging their natural role in transporting NA and protein-based cargo in cell-to-cell signaling. Their synthetic counterparts, lipid nanoparticles (LNPs), have been developed over the past decades as NA carriers, culminating in the approval of several marketed formulations such as patisiran/Onpattro® and the mRNA-1273/BNT162 COVID-19 vaccines. The success of LNPs has sparked efforts to develop innovative technologies to target extrahepatic organs, and to deliver novel therapeutic modalities, such as tools for in vivo gene editing. Fueled by the recent advancements in both fields, this review aims to provide a comprehensive overview of the basic characteristics of EV and LNP-based NA delivery systems, from EV biogenesis to structural properties of LNPs. It addresses the primary challenges encountered in utilizing these nanocarriers from a drug formulation and delivery perspective. Additionally, biodistribution profiles, in vitro and in vivo transfection outcomes, as well as their status in clinical trials are compared. Overall, this review provides insights into promising research avenues and potential dead ends for EV and LNP-based NA delivery systems.
Collapse
Affiliation(s)
- Johannes Bader
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, 8093 Zurich, Switzerland
| | - Finn Brigger
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, 8093 Zurich, Switzerland
| | - Jean-Christophe Leroux
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, 8093 Zurich, Switzerland.
| |
Collapse
|
26
|
Inano S, Kitano T. A modified CD9 tag for efficient protein delivery via extracellular vesicles. PLoS One 2024; 19:e0310083. [PMID: 39418272 PMCID: PMC11486436 DOI: 10.1371/journal.pone.0310083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Accepted: 08/23/2024] [Indexed: 10/19/2024] Open
Abstract
Extracellular vesicles (EVs) are attracting growing attention for therapeutic use and as diagnostic markers, particularly for cancer. Although therapies based on small interfering RNAs are under intensive research, other therapeutic molecules, especially proteins, have not been sufficiently investigated. One of the major method for loading proteins into EVs is electroporation; however, it damages membrane integrity and requires repeated purification, precluding clinical applications. Thus, natural and efficient protein transfer is a prerequisite for the clinical application of protein-based EV therapy. Another prerequisite is an efficient endosomal escape, as most EVs incorporated into receptor cells result in endosomal degradation. Therefore, we generated a short CD9 (sCD9)-INF/TAT tag for efficiently transfers fused proteins to the EV and enhances endosomal escape to address the abovementioned problems. Interestingly, protein transfer via EVs drastically improved when the EV producer and receptor cells were cocultured, strongly indicating bystander effects of cells producing therapeutic proteins fused with a sCD9-INF/TAT tag. This method can be applied to a wide range of therapeutic technologies, including cellular transplantation or viral therapy.
Collapse
Affiliation(s)
- Shojiro Inano
- Department of Hematology, Kitano Hospital, Osaka, Japan
- Kansai Electric Power Medical Research Institute, Osaka, Japan
- Kyoto Innovation Center for Next Generation Clinical Trials and iPS Cell Therapy (Ki-CONNECT), Kyoto University Hospital, Kyoto, Japan
| | | |
Collapse
|
27
|
Ikezu T, Yang Y, Verderio C, Krämer-Albers EM. Extracellular Vesicle-Mediated Neuron-Glia Communications in the Central Nervous System. J Neurosci 2024; 44:e1170242024. [PMID: 39358029 PMCID: PMC11450539 DOI: 10.1523/jneurosci.1170-24.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 07/17/2024] [Accepted: 07/24/2024] [Indexed: 10/04/2024] Open
Abstract
Communication between neurons and glia significantly influences the development maturation, plasticity, and disease progressions of the nervous system. As a new signaling modality, extracellular vesicles display a diverse role for robust functional regulation of neurons through their protein and nucleic acid cargoes. This review highlights recent breakthroughs in the research of signaling mechanisms between glia and neurons mediated by extracellular vesicles that are important for neural development, axonal maintenance, synaptic functions, and disease progression in the mammalian nervous system. We will discuss the biological roles of extracellular vesicles released from neurons, astroglia, microglia, and oligodendroglia in the nervous system and their implications in neurodegenerative disorders.
Collapse
Affiliation(s)
- Tsuneya Ikezu
- Department of Neuroscience, Mayo Clinic Florida, Jacksonville, Florida 32224
| | - Yongjie Yang
- Department of Neuroscience, Tufts University School of Medicine, Boston, Massachusetts 02111
| | - Claudia Verderio
- Department of Biomedical Sciences, CNR Institute of Neuroscience, Università Milano-Bicocca, 20854 Vedano al Lambro (MB), Italy
| | - Eva-Maria Krämer-Albers
- Institute of Developmental Biology and Neurobiology, Johannes Gutenberg University Mainz, 55128 Mainz, Rhineland Palatinate, Germany
| |
Collapse
|
28
|
Xia B, Hu R, Chen J, Shan S, Xu F, Zhang G, Zhou Z, Fan Y, Hu Z, Liang XJ. Oral Administration Properties Evaluation of Three Milk-Derived Extracellular Vesicles Based on Ultracentrifugation Extraction Methods. Adv Healthc Mater 2024; 13:e2401370. [PMID: 38767497 DOI: 10.1002/adhm.202401370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 05/16/2024] [Indexed: 05/22/2024]
Abstract
Milk-derived extracellular vesicles (M-EVs) are low-cost, can be prepared in large quantities, and can cross the gastrointestinal barrier for oral administration. However, the composition of milk is complex, and M-EVs obtained by different extraction methods may affect their oral delivery. Based on this, a new method for extracting M-EVs based on cryogenic freezing treatment (Cryo-M-EVs) is proposed and compared with the previously reported acetic acid treatment (Acid-M-EVs) method and the conventional ultracentrifugation method (Ulltr-M-EVs). The new method simplifies the pretreatment step and achieves 25-fold and twofold higher yields than Acid-M-EVs and Ulltr-M-EVs. And it is interesting to note that Cryo-M-EVs and Acid-M-EVs have higher cellular uptake efficiency, and Cryo-M-EVs present the best transepithelial transport effect. After oral administration of the three M-EVs extracted by three methods in mice, Cryo-M-EVs effectively successfully cross the gastrointestinal barrier and achieve hepatic accumulation, whereas Acid-M-EVs and Ultr-M-EVs mostly reside in the intestine. The M-EVs obtained by the three extraction methods show a favorable safety profile at the cellular as well as animal level. Therefore, when M-EVs obtained by different extraction methods are used for oral drug delivery, their accumulation properties at different sites can be utilized to better deal with different diseases.
Collapse
Affiliation(s)
- Bozhang Xia
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, No. 11, First North Road, Zhongguancun, Beijing, 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Runjing Hu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, No. 11, First North Road, Zhongguancun, Beijing, 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Junge Chen
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine & Shenzhen Institute of Beihang University, Beihang University, Beijing, 100083, China
| | - Shaobo Shan
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, No. 11, First North Road, Zhongguancun, Beijing, 100190, P. R. China
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100050, P. R. China
| | - Fengfei Xu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, No. 11, First North Road, Zhongguancun, Beijing, 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Gang Zhang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, No. 11, First North Road, Zhongguancun, Beijing, 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Ziran Zhou
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, No. 11, First North Road, Zhongguancun, Beijing, 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Yubo Fan
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine & Shenzhen Institute of Beihang University, Beihang University, Beijing, 100083, China
| | - Zhongbo Hu
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Xing-Jie Liang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, No. 11, First North Road, Zhongguancun, Beijing, 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| |
Collapse
|
29
|
Matloob A, Gu X, Rehman Sheikh A, Javed M, Fang Z, Luo Z. Plant exosomes‐like nano‐vesicles: Characterization, functional food potential, and emerging therapeutic applications as a nano medicine. FOOD SAFETY AND HEALTH 2024; 2:429-450. [DOI: 10.1002/fsh3.12060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 07/22/2024] [Indexed: 01/05/2025]
Abstract
AbstractPlant cells release exosome‐like nanovesicles (PENVs), which are small, membrane‐bound vesicles secreted by cells for intercellular interactions. These vesicles, rich in biologically active substances, are crucial for information transmission, intercellular interaction, and organism homeostasis conservation. They can also be used for treating diseases as large‐scale drug carriers due to their vesicular architecture. This study explores the isolation, potential of nanovesicles in creating bio‐therapeutic and drug‐delivery nano‐platforms to address clinical challenges. The bio‐therapeutic roles of PENVs, include immunomodulation, antitumor, regenerative impacts, wound healing, anti‐fibrosis, whitening effects, and treatment of intestinal flora disorders. This study also deliberates the potential for designing these nanovesicles into effective, safe, and non‐immunogenic nano‐vectors to carry drugs. PENVs may offer a cutting‐edge platform for the creation of functional foods and nutraceuticals. They might be employed to encapsulate certain bioactive substances produced from plants, offering tailored health privileges. It also elucidates the potential for the development of therapeutic and provision nano‐platforms based on PENVs in clinical applications.
Collapse
Affiliation(s)
- Anam Matloob
- College of Biosystems Engineering and Food Science Zhejiang University Hangzhou China
| | - Xinya Gu
- College of Biosystems Engineering and Food Science Zhejiang University Hangzhou China
| | - Arooj Rehman Sheikh
- College of Biosystems Engineering and Food Science Zhejiang University Hangzhou China
| | - Miral Javed
- College of Biosystems Engineering and Food Science Zhejiang University Hangzhou China
| | - Zhongxiang Fang
- School of Agriculture, Food and Ecosystem Sciences Faculty of Science The University of Melbourne Melbourne Victoria Australia
| | - Zisheng Luo
- College of Biosystems Engineering and Food Science Zhejiang University Hangzhou China
- Key Laboratory of Ago‐Products Postharvest Handing of Ministry of Agriculture and Rural Affairs Hangzhou China
| |
Collapse
|
30
|
Geng T, Tian L, Paek SY, Leung E, Chamley LW, Wu Z. Characterizing Extracellular Vesicles Generated from the Integra CELLine Culture System and Their Endocytic Pathways for Intracellular Drug Delivery. Pharmaceutics 2024; 16:1206. [PMID: 39339242 PMCID: PMC11434853 DOI: 10.3390/pharmaceutics16091206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Revised: 09/04/2024] [Accepted: 09/06/2024] [Indexed: 09/30/2024] Open
Abstract
Extracellular vesicles (EVs) have attracted great attention as promising intracellular drug delivery carriers. While the endocytic pathways of small EVs (sEVs, <200 nm) have been reported, there is limited understanding of large EVs (lEVs, >200 nm), despite their potential applications for drug delivery. Additionally, the low yield of EVs during isolation remains a major challenge in their application. Herein, we aimed to compare the endocytic pathways of sEVs and lEVs using MIA PaCa-2 pancreatic cancer cell-derived EVs as models and to explore the efficiency of their production. The cellular uptake of EVs by MIA PaCa-2 cells was assessed and the pathways were investigated with the aid of endocytic inhibitors. The yield and protein content of sEVs and lEVs from the Integra CELLine culture system and the conventional flasks were compared. Our findings revealed that both sEVs and lEVs produced by the Integra CELLine system entered their parental cells via multiple routes, including caveolin-mediated endocytosis, clathrin-mediated endocytosis, and actin-dependent phagocytosis or macropinocytosis. Notably, caveolin- and clathrin-mediated endocytosis were more prominent in the uptake of sEVs, while actin-dependent phagocytosis and macropinocytosis were significant for both sEVs and lEVs. Compared with conventional flasks, the Integra CELLine system demonstrated a 9-fold increase in sEVs yield and a 6.5-fold increase in lEVs yield, along with 3- to 4-fold higher protein content per 1010 EVs. Given that different endocytic pathways led to distinct intracellular trafficking routes, this study highlights the unique potentials of sEVs and lEVs for intracellular cargo delivery. The Integra CELLine proves to be a highly productive and cost-effective system for generating EVs with favourable properties for drug delivery.
Collapse
Affiliation(s)
- Tianjiao Geng
- School of Pharmacy, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1010, New Zealand; (T.G.); (L.T.)
- Department of Pharmacy, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Lei Tian
- School of Pharmacy, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1010, New Zealand; (T.G.); (L.T.)
| | - Song Yee Paek
- Department of Obstetrics and Gynaecology, Hub for Extracellular Vesicles Investigations, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1010, New Zealand; (S.Y.P.); (L.W.C.)
| | - Euphemia Leung
- Auckland Cancer Society Research Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1010, New Zealand;
| | - Lawrence W. Chamley
- Department of Obstetrics and Gynaecology, Hub for Extracellular Vesicles Investigations, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1010, New Zealand; (S.Y.P.); (L.W.C.)
| | - Zimei Wu
- School of Pharmacy, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1010, New Zealand; (T.G.); (L.T.)
| |
Collapse
|
31
|
Liu Y, Yin R, Tian Y, Xu S, Meng X. Curcumin nanopreparations: recent advance in preparation and application. Biomed Mater 2024; 19:052009. [PMID: 39189065 DOI: 10.1088/1748-605x/ad6dc7] [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: 04/15/2024] [Accepted: 08/09/2024] [Indexed: 08/28/2024]
Abstract
Curcumin is a natural polyphenolic compound extracted from turmeric with antibacterial, antioxidant, antitumor, preventive and therapeutic neurological disorders and a variety of bioactivities, which is widely used in the field of food and medicine. However, the drawbacks of curcumin such as poor aqueous solubility and stability have limited the practical application of curcumin. To overcome these defects and enhance its functional properties, various nanoscale systems (liposomes, polymer nanoparticles, protein nanoparticles, solid lipid nanoparticles, metal nanoparticles, etc) have been extensively employed for curcumin encapsulation and delivery. Despite the rapid development of curcumin nanoformulations, there is a lack of comprehensive reviews on their preparation and properties. This review provides an overview of the construction of curcumin nano-delivery systems, mechanisms of action, nanocarrier preparation methods and the applications of curcumin nanocarriers in the food and pharmaceutical fields to provide a theoretical basis and technological support for the efficient bio-utilization, product development and early clinical application of curcumin.
Collapse
Affiliation(s)
- Yan Liu
- School of Pharmacy, Heilongjiang University of Chinese Medicine, NO.24 Heping Road, Harbin, 150040, People's Republic of China
| | - Rui Yin
- School of Pharmacy, Heilongjiang University of Chinese Medicine, NO.24 Heping Road, Harbin, 150040, People's Republic of China
| | - Yuan Tian
- School of Pharmacy, Heilongjiang University of Chinese Medicine, NO.24 Heping Road, Harbin, 150040, People's Republic of China
| | - Shujun Xu
- School of Pharmacy, Heilongjiang University of Chinese Medicine, NO.24 Heping Road, Harbin, 150040, People's Republic of China
| | - Xin Meng
- School of Pharmacy, Heilongjiang University of Chinese Medicine, NO.24 Heping Road, Harbin, 150040, People's Republic of China
| |
Collapse
|
32
|
Xiang H, Bao C, Chen Q, Gao Q, Wang N, Gao Q, Mao L. Extracellular vesicles (EVs)' journey in recipient cells: from recognition to cargo release. J Zhejiang Univ Sci B 2024; 25:633-655. [PMID: 39155778 PMCID: PMC11337091 DOI: 10.1631/jzus.b2300566] [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: 08/13/2023] [Accepted: 11/28/2023] [Indexed: 08/20/2024]
Abstract
Extracellular vesicles (EVs) are nano-sized bilayer vesicles that are shed or secreted by virtually every cell type. A variety of biomolecules, including proteins, lipids, coding and non-coding RNAs, and mitochondrial DNA, can be selectively encapsulated into EVs and delivered to nearby and distant recipient cells, leading to alterations in the recipient cells, suggesting that EVs play an important role in intercellular communication. EVs play effective roles in physiology and pathology and could be used as diagnostic and therapeutic tools. At present, although the mechanisms of exosome biogenesis and secretion in donor cells are well understood, the molecular mechanism of EV recognition and uptake by recipient cells is still unclear. This review summarizes the current understanding of the molecular mechanisms of EVs' biological journey in recipient cells, from recognition to uptake and cargo release. Furthermore, we highlight how EVs escape endolysosomal degradation after uptake and thus release cargo, which is crucial for studies applying EVs as drug-targeted delivery vehicles. Knowledge of the cellular processes that govern EV uptake is important to shed light on the functions of EVs as well as on related clinical applications.
Collapse
Affiliation(s)
- Huayuan Xiang
- Department of Laboratory Medicine, Affiliated Kunshan Hospital of Jiangsu University, Kunshan 215300, China
| | - Chenxuan Bao
- Department of Laboratory Medicine, Affiliated Kunshan Hospital of Jiangsu University, Kunshan 215300, China
| | - Qiaoqiao Chen
- Department of Laboratory Medicine, Affiliated Kunshan Hospital of Jiangsu University, Kunshan 215300, China
| | - Qing Gao
- Department of Laboratory Medicine, Affiliated Kunshan Hospital of Jiangsu University, Kunshan 215300, China
| | - Nan Wang
- Department of Laboratory Medicine, Affiliated Kunshan Hospital of Jiangsu University, Kunshan 215300, China
| | - Qianqian Gao
- Department of Laboratory Medicine, Affiliated Kunshan Hospital of Jiangsu University, Kunshan 215300, China
| | - Lingxiang Mao
- Department of Laboratory Medicine, Affiliated Kunshan Hospital of Jiangsu University, Kunshan 215300, China.
| |
Collapse
|
33
|
Morishita M, Makabe M, Shinohara C, Fukumori A, Morita S, Terada Y, Miyai S, Katsumi H, Yamamoto A. Versatile functionalization of Bifidobacteria-derived extracellular vesicles using amino acid metabolic labeling and click chemistry for immunotherapy. Int J Pharm 2024; 661:124410. [PMID: 38954931 DOI: 10.1016/j.ijpharm.2024.124410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Revised: 06/02/2024] [Accepted: 06/29/2024] [Indexed: 07/04/2024]
Abstract
Extracellular vesicles (EVs) are nanoparticles secreted by various organisms. Methods for modifying EVs functionally have garnered attention for developing EV-based therapeutic systems. However, most technologies used to integrate these functions are limited to mammalian-derived EVs and a promising modification method for bacteria-derived EVs has not yet been developed. In this study, we propose a novel method for the versatile functionalization of immunostimulatory probiotic Bifidobacteria-derived EVs (B-EVs) using amino acid metabolic labeling and azide-alkyne click reaction. Azide D-alanine (ADA), a similar molecule to D-alanine in bacteria cell-wall peptidoglycan, was selected as an azide group-functionalized amino acid. Azide-modified B-EVs were isolated from Bifidobacteria incubated with ADA. The physicochemical and compositional characteristics, as well as adjuvanticity of B-EVs against immune cells were not affected by azide loading, demonstrating that this functionalization approach can retain the endogenous usefulness of B-EVs. By using the fluorescent B-EVs obtained by this method, the intracellular trafficking of B-EVs after uptake by immune cells was successfully observed. Furthermore, this method enabled the formulation of B-EVs for hydrogelation and enhanced adjuvanticity in the host. Our findings will be helpful for further development of EV-based immunotherapy.
Collapse
Affiliation(s)
- Masaki Morishita
- Department of Biopharmaceutics, Kyoto Pharmaceutical University, Misasagi, Yamashina-Ku, Kyoto 607-8414, Japan.
| | - Mizuho Makabe
- Department of Biopharmaceutics, Kyoto Pharmaceutical University, Misasagi, Yamashina-Ku, Kyoto 607-8414, Japan
| | - Chisa Shinohara
- Department of Biopharmaceutics, Kyoto Pharmaceutical University, Misasagi, Yamashina-Ku, Kyoto 607-8414, Japan
| | - Ami Fukumori
- Department of Biopharmaceutics, Kyoto Pharmaceutical University, Misasagi, Yamashina-Ku, Kyoto 607-8414, Japan
| | - Shiori Morita
- Department of Biopharmaceutics, Kyoto Pharmaceutical University, Misasagi, Yamashina-Ku, Kyoto 607-8414, Japan
| | - Yuki Terada
- Department of Biopharmaceutics, Kyoto Pharmaceutical University, Misasagi, Yamashina-Ku, Kyoto 607-8414, Japan
| | - Syunsuke Miyai
- Department of Biopharmaceutics, Kyoto Pharmaceutical University, Misasagi, Yamashina-Ku, Kyoto 607-8414, Japan
| | - Hidemasa Katsumi
- Department of Biopharmaceutics, Kyoto Pharmaceutical University, Misasagi, Yamashina-Ku, Kyoto 607-8414, Japan
| | - Akira Yamamoto
- Department of Biopharmaceutics, Kyoto Pharmaceutical University, Misasagi, Yamashina-Ku, Kyoto 607-8414, Japan
| |
Collapse
|
34
|
Tang M, Yarragudi SB, Pan P, Yang K, Kanamala M, Wu Z. Effect of size and pH-sensitivity of liposomes on cellular uptake pathways and pharmacokinetics of encapsulated gemcitabine. J Liposome Res 2024:1-11. [PMID: 39126197 DOI: 10.1080/08982104.2024.2389969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 08/02/2024] [Accepted: 08/03/2024] [Indexed: 08/12/2024]
Abstract
To enhance cytoplasmic delivery efficiency, pH-sensitive liposomes (PSL) have been proposed as a novel strategy. To facilitate clinical translation, this study aims to understand the impact of both size and pH-sensitivity on cellular uptake pathways, intracellular trafficking and pharmacokinetics of liposomes. The large liposomes (130-160 nm) were prepared using thin-film hydration method, while small liposomes (∼60 nm) were fabricated using microfluidics, for both PSL and non-pH-sensitive liposomes (NPSL). Cellular uptake pathways and intracellular trafficking was investigated through confocal imaging with aid of various endocytosis inhibitors. Intracellular gemcitabine delivery by various liposomal formulations was quantified using HPLC, and the cytotoxicity was assessed via cell viability assays. Pharmacokinetics of gemcitabine loaded in various liposomes was evaluated in rats following intravenous administration. Larger liposomes had a higher loading capacity for hydrophilic gemcitabine (7% vs 4%). Small PSL exhibited superior cellular uptake compared to large PSL or NPSLs. Moreover, the alkalization of endosomes significantly attenuated the cellular uptake of PSL. Large liposomes (PSL and NPSL) predominantly entered cells via clathrin-dependent pathway, whereas small liposomes partially utilized caveolae-dependent pathway. However, the long circulation of the liposomes, as measured by the encapsulated gemcitabine, was compromised by both pH-sensitivity and size reduction (9.5 h vs 5.3 h). Despite this drawback, our results indicate that small PSL holds promise as vectors for the next generation of liposomal nanomedicine, owing to their superior cytoplasmic delivery efficiency.
Collapse
Affiliation(s)
- Mingtan Tang
- School of Pharmacy, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, China
| | - Sasi Bhushan Yarragudi
- School of Pharmacy, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Patrick Pan
- School of Pharmacy, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Kaiyun Yang
- School of Pharmacy, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Manju Kanamala
- School of Pharmacy, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Zimei Wu
- School of Pharmacy, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| |
Collapse
|
35
|
Wang J, Yin B, Lian J, Wang X. Extracellular Vesicles as Drug Delivery System for Cancer Therapy. Pharmaceutics 2024; 16:1029. [PMID: 39204374 PMCID: PMC11359799 DOI: 10.3390/pharmaceutics16081029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 07/18/2024] [Accepted: 07/29/2024] [Indexed: 09/04/2024] Open
Abstract
In recent decades, the pursuit of drug delivery systems has led to the development of numerous synthetic options aimed at enhancing drug efficacy while minimizing side effects. However, the practical application of these systems is often hindered by challenges such as inefficiency, cytotoxicity, and immunogenicity. Extracellular vesicles, natural carriers for drugs, emerge as promising alternatives with distinct advantages over synthetic carriers. Notably, EVs exhibit biocompatibility, low immunogenicity, and inherent tissue-targeting capabilities, thus opening new avenues for drug delivery strategies. This review provides an overview of EVs, including their biogenesis and absorption mechanisms. Additionally, we explore the current research efforts focusing on harnessing their potential as drug carriers, encompassing aspects such as purification techniques, drug loading, and bioengineering for targeted delivery. Finally, we discuss the existing challenges and future prospects of EVs as therapeutic agents in clinical settings. This comprehensive analysis aims to shed light on the potential of EVs as versatile and effective tools for drug delivery, particularly in the realm of cancer therapy.
Collapse
Affiliation(s)
- Jin Wang
- School of Life Sciences, Liaoning University, Shenyang 110036, China; (J.W.); (J.L.)
| | - Bohang Yin
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University, Shenyang 110001, China;
| | - Jiabing Lian
- School of Life Sciences, Liaoning University, Shenyang 110036, China; (J.W.); (J.L.)
| | - Xia Wang
- Institute of Health Sciences, China Medical University, 77 Puhe Road, Shenyang 110122, China
| |
Collapse
|
36
|
Rahimian S, Najafi H, Webber CA, Jalali H. Advances in Exosome-Based Therapies for the Repair of Peripheral Nerve Injuries. Neurochem Res 2024; 49:1905-1925. [PMID: 38807021 DOI: 10.1007/s11064-024-04157-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 03/07/2024] [Accepted: 05/17/2024] [Indexed: 05/30/2024]
Abstract
Peripheral nerve injuries (PNIs) are the term used to describe injuries that occur to the nerve fibers of the peripheral nervous system (PNS). Such injuries may be caused by trauma, infection, or aberrant immunological response. Although the peripheral nervous system has a limited capacity for self-repair, in cases of severe damage, this process is either interrupted entirely or is only partially completed. The evaluation of variables that promote the repair of peripheral nerves has consistently been a focal point. Exosomes are a subtype of extracellular vesicles that originate from cellular sources and possess abundant proteins, lipids, and nucleic acids, play a critical role in facilitating intercellular communication. Due to their modifiable composition, they possess exceptional capabilities as carriers for therapeutic compounds, including but not limited to mRNAs or microRNAs. Exosome-based therapies have gained significant attention in the treatment of several nervous system diseases due to their advantageous properties, such as low toxicity, high stability, and limited immune system activation. The objective of this review article is to provide an overview of exosome-based treatments that have been developed in recent years for a range of PNIs, including nerve trauma, diabetic neuropathy, amyotrophic lateral sclerosis (ALS), glaucoma, and Guillain-Barre syndrome (GBS). It was concluded that exosomes could provide favorable results in the improvement of peripheral PNIs by facilitating the transfer of regenerative factors. The development of bioengineered exosome therapy for PNIs should be given more attention to enhance the efficacy of exosome treatment for PNIs.
Collapse
Affiliation(s)
- Sana Rahimian
- Division of Nanobiotehnology, Department of Cell and Molecular Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | - Hossein Najafi
- Division of Nanobiotehnology, Department of Cell and Molecular Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | - Christine A Webber
- Division of Anatomy, Department of Surgery, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Hanieh Jalali
- Division of Cell and Developmental Biology, Department of Animal Biology, Faculty of Biological Sciences, Kharazmi University, No. 43, South Moffateh Ave, Tehran, 15719-14911, Iran.
| |
Collapse
|
37
|
Yadav K, Gnanakani SPE, Sahu KK, Veni Chikkula CK, Vaddi PS, Srilakshmi S, Yadav R, Sucheta, Dubey A, Minz S, Pradhan M. Nano revolution of DNA nanostructures redefining cancer therapeutics-A comprehensive review. Int J Biol Macromol 2024; 274:133244. [PMID: 38901506 DOI: 10.1016/j.ijbiomac.2024.133244] [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: 01/10/2024] [Revised: 06/10/2024] [Accepted: 06/16/2024] [Indexed: 06/22/2024]
Abstract
DNA nanostructures are a promising tool in cancer treatment, offering an innovative way to improve the effectiveness of therapies. These nanostructures can be made solely from DNA or combined with other materials to overcome the limitations of traditional single-drug treatments. There is growing interest in developing nanosystems capable of delivering multiple drugs simultaneously, addressing challenges such as drug resistance. Engineered DNA nanostructures are designed to precisely deliver different drugs to specific locations, enhancing therapeutic effects. By attaching targeting molecules, these nanostructures can recognize and bind to cancer cells, increasing treatment precision. This approach offers tailored solutions for targeted drug delivery, enabling the delivery of multiple drugs in a coordinated manner. This review explores the advancements and applications of DNA nanostructures in cancer treatment, with a focus on targeted drug delivery and multi-drug therapy. It discusses the benefits and current limitations of nanoscale formulations in cancer therapy, categorizing DNA nanostructures into pure forms and hybrid versions optimized for drug delivery. Furthermore, the review examines ongoing research efforts and translational possibilities, along with challenges in clinical integration. By highlighting the advancements in DNA nanostructures, this review aims to underscore their potential in improving cancer treatment outcomes.
Collapse
Affiliation(s)
- Krishna Yadav
- Rungta College of Pharmaceutical Sciences and Research, Kohka, Bhilai 490024, India
| | - S Princely E Gnanakani
- Department of Pharmaceutical Biotechnology, Parul Institute of Pharmacy, Parul University, Post Limda, Ta.Waghodia - 391760, Dist. Vadodara, Gujarat, India
| | - Kantrol Kumar Sahu
- Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh 281406, India
| | - C Krishna Veni Chikkula
- Department of Environmental Toxicology, Southern University and A&M College, Baton Rouge, LA, USA
| | - Poorna Sai Vaddi
- Department of Environmental Toxicology, Southern University and A&M College, Baton Rouge, LA, USA
| | - S Srilakshmi
- Gitam School of Pharmacy, Department of Pharmaceutical Chemistry, Gitams University, Vishakhapatnam, India
| | - Renu Yadav
- School of Medical and Allied Sciences, K. R. Mangalam University, Sohna Road, Gurugram, Haryana 122103, India
| | - Sucheta
- School of Medical and Allied Sciences, K. R. Mangalam University, Sohna Road, Gurugram, Haryana 122103, India
| | - Akhilesh Dubey
- Nitte (Deemed to be University), NGSM Institute of Pharmaceutical Sciences, Department of Pharmaceutics, Mangaluru 575018, Karnataka, India
| | - Sunita Minz
- Department of Pharmacy, Indira Gandhi National Tribal University, Amarkantak (M.P.), India
| | | |
Collapse
|
38
|
Huang S, Lin J, Han X. Extracellular vesicles-Potential link between periodontal disease and diabetic complications. Mol Oral Microbiol 2024; 39:225-239. [PMID: 38227219 DOI: 10.1111/omi.12449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 12/06/2023] [Accepted: 12/25/2023] [Indexed: 01/17/2024]
Abstract
It has long been suggested that a bidirectional impact exists between periodontitis and diabetes. Periodontitis may affect diabetes glycemic control, insulin resistance, and diabetic complications. Diabetes can worsen periodontitis by delaying wound healing and increasing the chance of infection. Extracellular vesicles (EVs) are heterogeneous particles of membrane-enclosed spherical structure secreted by eukaryotes and prokaryotes and play a key role in a variety of diseases. This review will introduce the biogenesis, release, and biological function of EVs from a microbial and host cell perspective, discuss the functional properties of EVs in the development of periodontitis and diabetes, and explore their role in the pathogenesis and clinical application of these two diseases. Their clinical implication and diagnostic value are also discussed.
Collapse
Affiliation(s)
- Shengyuan Huang
- Department of Oral Science and Translation Research, College of Dental Medicine, Nova Southeastern University, Fort Lauderdale, Florida, USA
- Department of Stomatology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Jiang Lin
- Department of Stomatology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Xiaozhe Han
- Department of Oral Science and Translation Research, College of Dental Medicine, Nova Southeastern University, Fort Lauderdale, Florida, USA
| |
Collapse
|
39
|
Licari E, Cricrì G, Mauri M, Raimondo F, Dioni L, Favero C, Giussani A, Starace R, Nucera S, Biondi A, Piazza R, Bollati V, Dander E, D'Amico G. ActivinA modulates B-acute lymphoblastic leukaemia cell communication and survival by inducing extracellular vesicles production. Sci Rep 2024; 14:16083. [PMID: 38992199 PMCID: PMC11239915 DOI: 10.1038/s41598-024-66779-3] [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: 03/13/2024] [Accepted: 07/03/2024] [Indexed: 07/13/2024] Open
Abstract
Extracellular vesicles (EVs) are a new mechanism of cellular communication, by delivering their cargo into target cells to modulate molecular pathways. EV-mediated crosstalk contributes to tumor survival and resistance to cellular stress. However, the role of EVs in B-cell Acute Lymphoblastic Leukaemia (B-ALL) awaits to be thoroughly investigated. We recently published that ActivinA increases intracellular calcium levels and promotes actin polymerization in B-ALL cells. These biological processes guide cytoskeleton reorganization, which is a crucial event for EV secretion and internalization. Hence, we investigated the role of EVs in the context of B-ALL and the impact of ActivinA on this phenomenon. We demonstrated that leukemic cells release a higher number of EVs in response to ActivinA treatment, and they can actively uptake EVs released by other B-ALL cells. Under culture-induced stress conditions, EVs coculture promoted cell survival in B-ALL cells in a dose-dependent manner. Direct stimulation of B-ALL cells with ActivinA or with EVs isolated from ActivinA-stimulated cells was even more effective in preventing cell death. This effect can be possibly ascribed to the increase of vesiculation and modifications of EV-associated microRNAs induced by ActivinA. These data demonstrate that ActivinA boosts EV-mediated B-ALL crosstalk, improving leukemia survival in stress conditions.
Collapse
Affiliation(s)
- Eugenia Licari
- Tettamanti Center, Fondazione IRCCS San Gerardo dei Tintori, Via Pergolesi, 20900, Monza, Italy
| | - Giulia Cricrì
- Tettamanti Center, Fondazione IRCCS San Gerardo dei Tintori, Via Pergolesi, 20900, Monza, Italy
- Paediatric Nephrology, Dialysis and Transplant Unit, Fondazione Ca' Granda IRCCS Ospedale Maggiore Policlinico, Milano, Italy
| | - Mario Mauri
- Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Francesca Raimondo
- Clinical Proteomics and Metabolomic Unit, School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Laura Dioni
- EPIGET Lab, Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Chiara Favero
- EPIGET Lab, Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Alice Giussani
- Tettamanti Center, Fondazione IRCCS San Gerardo dei Tintori, Via Pergolesi, 20900, Monza, Italy
| | - Rita Starace
- Tettamanti Center, Fondazione IRCCS San Gerardo dei Tintori, Via Pergolesi, 20900, Monza, Italy
| | - Silvia Nucera
- Tettamanti Center, Fondazione IRCCS San Gerardo dei Tintori, Via Pergolesi, 20900, Monza, Italy
- Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Andrea Biondi
- Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
- Pediatrics, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | - Rocco Piazza
- Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
- Hematology Division and Bone Marrow Unit, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | - Valentina Bollati
- EPIGET Lab, Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
- CRC, Center for Environmental Health, University of Milan, Milan, Italy
- Occupational Health Unit, Fondazione IRCCS Ca' Granda-Ospedale Maggiore Policlinico, Milan, Italy
| | - Erica Dander
- Tettamanti Center, Fondazione IRCCS San Gerardo dei Tintori, Via Pergolesi, 20900, Monza, Italy
| | - Giovanna D'Amico
- Tettamanti Center, Fondazione IRCCS San Gerardo dei Tintori, Via Pergolesi, 20900, Monza, Italy.
| |
Collapse
|
40
|
Wang S, Kong H, Zhuo C, Liu L, Lv S, Cheng D, Lao YH, Tao Y, Li M. Functionalized extracellular nanovesicles as advanced CRISPR delivery systems. Biomater Sci 2024; 12:3480-3499. [PMID: 38808607 DOI: 10.1039/d4bm00054d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/30/2024]
Abstract
The clustered regularly interspaced short palindromic repeat (CRISPR) system, an emerging tool for genome editing, has garnered significant public interest for its potential in treating genetic diseases. Despite the rapid advancements in CRISPR technology, the progress in developing effective delivery strategies lags, impeding its clinical application. Extracellular nanovesicles (EVs), either in their endogenous forms or with engineered modifications, have emerged as a promising solution for CRISPR delivery. These EVs offer several advantages, including high biocompatibility, biological permeability, negligible immunogenicity, and straightforward production. Herein, we first summarize various types of functional EVs for CRISPR delivery, such as unmodified, modified, engineered virus-like particles (VLPs), and exosome-liposome hybrid vesicles, and examine their distinct intracellular pathways. Then, we outline the cutting-edge techniques for functionalizing extracellular vesicles, involving producer cell engineering, vesicle engineering, and virus-like particle engineering, emphasizing the diverse CRISPR delivery capabilities of these nanovesicles. Lastly, we address the current challenges and propose rational design strategies for their clinical translation, offering future perspectives on the development of functionalized EVs.
Collapse
Affiliation(s)
- Siqing Wang
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China.
| | - Huimin Kong
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China.
| | - Chenya Zhuo
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China.
| | - Li Liu
- Department of Gynecology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518000, China
| | - Shixian Lv
- School of Materials Science and Engineering, Peking University, Beijing, 100871, China
| | - Du Cheng
- PCFM Lab of Ministry of Education, School of Material Science and Engineering, Sun Yat-Sen University, Guangzhou 510275, China
| | - Yeh-Hsing Lao
- Department of Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, NY 14214, USA.
| | - Yu Tao
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China.
| | - Mingqiang Li
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China.
- Guangdong Provincial Key Laboratory of Liver Disease Research, Guangzhou 510630, China
| |
Collapse
|
41
|
Wang S, Qiao C, Kong X, Yang J, Guo F, Chen J, Wang W, Zhang B, Xiu H, He Y, Wang J, Feng H, Cai Z. Adhesion between EVs and tumor cells facilitated EV-encapsulated doxorubicin delivery via ICAM1. Pharmacol Res 2024; 205:107244. [PMID: 38821149 DOI: 10.1016/j.phrs.2024.107244] [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: 03/20/2024] [Revised: 05/28/2024] [Accepted: 05/28/2024] [Indexed: 06/02/2024]
Abstract
Doxorubicin (Dox) is an anti-tumor drug with a broad spectrum, whereas the cardiotoxicity limits its further application. In clinical settings, liposome delivery vehicles are used to reduce Dox cardiotoxicity. Here, we substitute extracellular vesicles (EVs) for liposomes and deeply investigate the mechanism for EV-encapsulated Dox delivery. The results demonstrate that EVs dramatically increase import efficiency and anti-tumor effects of Dox in vitro and in vivo, and the efficiency increase benefits from its unique entry pattern. Dox-loading EVs repeat a "kiss-and-run" motion before EVs internalization. Once EVs touch the cell membrane, Dox disassociates from EVs and directly enters the cytoplasm, leading to higher and faster Dox import than single Dox. This unique entry pattern makes the adhesion between EVs and cell membrane rather than the total amount of EV internalization the key factor for regulating the Dox import. Furthermore, we recognize ICAM1 as the molecule mediating the adhesion between EVs and cell membranes. Interestingly, EV-encapsulated Dox can induce ICAM1 expression by irritating IFN-γ and TNF-α secretion in TME, thereby increasing tumor targeting of Dox-loading EVs. Altogether, EVs and EV-encapsulated Dox synergize via ICAM1, which collectively enhances the curative effects for tumor treatment.
Collapse
Affiliation(s)
- Shibo Wang
- Institute of Immunology and Department of Orthopaedics of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China; Institute of Immunology and Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China; Institute of Hematology, Zhejiang University & Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou 310006, China
| | - Chenxiao Qiao
- Institute of Immunology and Department of Orthopaedics of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Xianghui Kong
- Institute of Immunology and Department of Orthopaedics of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Jie Yang
- Department of Critical Care Medicine of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Fei Guo
- Department of Urology, Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Jiming Chen
- Institute of Immunology and Department of Orthopaedics of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Wenhui Wang
- Institute of Immunology and Department of Orthopaedics of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Bei Zhang
- Institute of Immunology and Department of Orthopaedics of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Huiqing Xiu
- Department of Critical Care Medicine of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Yuzhou He
- Department of Emergency, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310053, China.
| | - Jianli Wang
- Institute of Immunology and Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China; Institute of Hematology, Zhejiang University & Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou 310006, China.
| | - Huajun Feng
- Ecological-Environment & Health College, Zhejiang A & F University, Hangzhou, Zhejiang 311300, China.
| | - Zhijian Cai
- Institute of Immunology and Department of Orthopaedics of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China.
| |
Collapse
|
42
|
Fazeli A, Godakumara K. The evolving roles of extracellular vesicles in embryo-maternal communication. Commun Biol 2024; 7:754. [PMID: 38906986 PMCID: PMC11192758 DOI: 10.1038/s42003-024-06442-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 06/12/2024] [Indexed: 06/23/2024] Open
Abstract
Mammalian reproduction relies on precise maternal-fetal communication, wherein immune modifications foster tolerance toward the semi-allogeneic embryo. Extracellular vesicles (EVs), including exosomes and microvesicles, have emerged as crucial mediators, transporting molecules like microRNAs securely. EVs influence various reproductive stages, from gamete maturation to implantation, and impact pathologies like pregnancy loss. In the embryo-maternal dialogue, EVs notably affect oviductal interactions, gene expression, and the embryo-endometrial interface, crucial for successful implantation. Key queries persist about EV uptake, cargo delivery, and the specific biomolecules driving communication. Their potential in diagnostics, therapeutics, and understanding environmental impacts on fertility signals an exciting future, reliant on collaborative efforts for transformative strides in reproductive health.
Collapse
Affiliation(s)
- Alireza Fazeli
- Institute of Veterinary Medicine and Animal Sciences, Estonian University of Life Sciences, Tartu, Estonia.
- Department of Pathophysiology, Institute of Biomedicine and Translational Medicine, Faculty of Medicine, Tartu University, Tartu, Estonia.
- Division of Clinical Medicine, School of Medicine & Population Health, University of Sheffield, Sheffield, UK.
| | - Kasun Godakumara
- Institute of Veterinary Medicine and Animal Sciences, Estonian University of Life Sciences, Tartu, Estonia
| |
Collapse
|
43
|
Zhou H, Li Z, Jing S, Wang B, Ye Z, Xiong W, Liu Y, Liu Y, Xu C, Kumeria T, He Y, Ye Q. Repair spinal cord injury with a versatile anti-oxidant and neural regenerative nanoplatform. J Nanobiotechnology 2024; 22:351. [PMID: 38902789 PMCID: PMC11188197 DOI: 10.1186/s12951-024-02610-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 05/31/2024] [Indexed: 06/22/2024] Open
Abstract
Spinal cord injury (SCI) often results in motor and sensory deficits, or even paralysis. Due to the role of the cascade reaction, the effect of excessive reactive oxygen species (ROS) in the early and middle stages of SCI severely damage neurons, and most antioxidants cannot consistently eliminate ROS at non-toxic doses, which leads to a huge compromise in antioxidant treatment of SCI. Selenium nanoparticles (SeNPs) have excellent ROS scavenging bioactivity, but the toxicity control problem limits the therapeutic window. Here, we propose a synergistic therapeutic strategy of SeNPs encapsulated by ZIF-8 (SeNPs@ZIF-8) to obtain synergistic ROS scavenging activity. Three different spatial structures of SeNPs@ZIF-8 were synthesized and coated with ferrostatin-1, a ferroptosis inhibitor (FSZ NPs), to achieve enhanced anti-oxidant and anti-ferroptosis activity without toxicity. FSZ NPs promoted the maintenance of mitochondrial homeostasis, thereby regulating the expression of inflammatory factors and promoting the polarization of macrophages into M2 phenotype. In addition, the FSZ NPs presented strong abilities to promote neuronal maturation and axon growth through activating the WNT4-dependent pathways, while prevented glial scar formation. The current study demonstrates the powerful and versatile bioactive functions of FSZ NPs for SCI treatment and offers inspiration for other neural injury diseases.
Collapse
Affiliation(s)
- Heng Zhou
- Center of Regenerative Medicine, Department of Stomatology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Ziwei Li
- Center of Regenerative Medicine, Department of Stomatology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Shuili Jing
- Center of Regenerative Medicine, Department of Stomatology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Ben Wang
- The Second People's Hospital of Linhai, Linhai, Zhejiang, 317000, China
| | - Zhifei Ye
- The Second People's Hospital of Linhai, Linhai, Zhejiang, 317000, China
| | - Wei Xiong
- Center of Regenerative Medicine, Department of Stomatology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Yonghao Liu
- Center of Regenerative Medicine, Department of Stomatology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Ye Liu
- Center of Regenerative Medicine, Department of Stomatology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Chun Xu
- School of Dentistry, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Tushar Kumeria
- School of Materials Science and Engineering, University of New South Wales, Kensington, Sydney, NSW, 2052, Australia
| | - Yan He
- Institute of Regenerative and Translational Medicine, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, 430064, Hubei, China
- Department of Oral and Maxillofacial Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
| | - Qingsong Ye
- Center of Regenerative Medicine, Department of Stomatology, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
- The Second People's Hospital of Linhai, Linhai, Zhejiang, 317000, China.
| |
Collapse
|
44
|
Guerricchio L, Barile L, Bollini S. Evolving Strategies for Extracellular Vesicles as Future Cardiac Therapeutics: From Macro- to Nano-Applications. Int J Mol Sci 2024; 25:6187. [PMID: 38892376 PMCID: PMC11173118 DOI: 10.3390/ijms25116187] [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: 04/12/2024] [Revised: 05/28/2024] [Accepted: 05/31/2024] [Indexed: 06/21/2024] Open
Abstract
Cardiovascular disease represents the foremost cause of mortality and morbidity worldwide, with a steadily increasing incidence due to the growth of the ageing population. Cardiac dysfunction leading to heart failure may arise from acute myocardial infarction (MI) as well as inflammatory- and cancer-related chronic cardiomyopathy. Despite pharmacological progress, effective cardiac repair represents an unmet clinical need, with heart transplantation being the only option for end-stage heart failure. The functional profiling of the biological activity of extracellular vesicles (EVs) has recently attracted increasing interest in the field of translational research for cardiac regenerative medicine. The cardioprotective and cardioactive potential of human progenitor stem/cell-derived EVs has been reported in several preclinical studies, and EVs have been suggested as promising paracrine therapy candidates for future clinical translation. Nevertheless, some compelling aspects must be properly addressed, including optimizing delivery strategies to meet patient needs and enhancing targeting specificity to the cardiac tissue. Therefore, in this review, we will discuss the most relevant aspects of the therapeutic potential of EVs released by human progenitors for cardiovascular disease, with a specific focus on the strategies that have been recently implemented to improve myocardial targeting and administration routes.
Collapse
Affiliation(s)
- Laura Guerricchio
- Biology Unit, Department of Experimental Medicine (DIMES), University of Genova, 16132 Genova, Italy;
| | - Lucio Barile
- Cardiovascular Theranostics, Istituto Cardiocentro Ticino, Laboratories for Translational Research, Ente Ospedaliero Cantonale, CH-6500 Bellinzona, Switzerland;
- Euler Institute, Faculty of Biomedical Sciences, Università della Svizzera Italiana, CH-6900 Lugano, Switzerland
| | - Sveva Bollini
- Biology Unit, Department of Experimental Medicine (DIMES), University of Genova, 16132 Genova, Italy;
- Cellular Oncology Unit, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy
| |
Collapse
|
45
|
Ye Z, Chen W, Li G, Huang J, Lei J. Tissue-derived extracellular vesicles in cancer progression: mechanisms, roles, and potential applications. Cancer Metastasis Rev 2024; 43:575-595. [PMID: 37851319 DOI: 10.1007/s10555-023-10147-6] [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: 07/29/2023] [Accepted: 10/03/2023] [Indexed: 10/19/2023]
Abstract
Extracellular vesicles (EVs) are small lipid bilayer-enclosed vesicles that mediate vital cellular communication by transferring cargo between cells. Among these, tissue-derived extracellular vesicles (Ti-EVs) stand out due to their origin from the tissue microenvironment, providing a more accurate reflection of changes in this setting. This unique advantage makes Ti-EVs valuable in investigating the intricate relationship between extracellular vesicles and cancer progression. Despite considerable research efforts exploring the association between Ti-EVs and cancers, a comprehensive clustering or grouping of these studies remains lacking. In this review, we aim to fill this gap by presenting a comprehensive synthesis of the mechanisms underlying Ti-EV generation, release, and transport within cancer tissues. Moreover, we delve into the pivotal roles that Ti-EVs play in cancer progression, shedding light on their potential as diagnostic and therapeutic tools. The review culminates in the construction of a comprehensive functional spectrum of Ti-EVs, providing a valuable reference for future research endeavors. By summarizing the current state of knowledge on Ti-EVs and their significance in tumor biology, this work contributes to a deeper understanding of cancer microenvironment dynamics and opens up avenues for harnessing Ti-EVs in diagnostic and therapeutic applications.
Collapse
Affiliation(s)
- Ziyang Ye
- Division of Thyroid Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Wenjie Chen
- Division of Thyroid Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Genpeng Li
- Division of Thyroid Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jing Huang
- Division of Thyroid Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jianyong Lei
- Division of Thyroid Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
46
|
Wang Y, Ding S. Extracellular vesicles in cancer cachexia: deciphering pathogenic roles and exploring therapeutic horizons. J Transl Med 2024; 22:506. [PMID: 38802952 PMCID: PMC11129506 DOI: 10.1186/s12967-024-05266-9] [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: 03/12/2024] [Accepted: 04/29/2024] [Indexed: 05/29/2024] Open
Abstract
Cancer cachexia (CC) is a debilitating syndrome that affects 50-80% of cancer patients, varying in incidence by cancer type and significantly diminishing their quality of life. This multifactorial syndrome is characterized by muscle and fat loss, systemic inflammation, and metabolic imbalance. Extracellular vesicles (EVs), including exosomes and microvesicles, play a crucial role in the progression of CC. These vesicles, produced by cancer cells and others within the tumor environment, facilitate intercellular communication by transferring proteins, lipids, and nucleic acids. A comprehensive review of the literature from databases such as PubMed, Scopus, and Web of Science reveals insights into the formation, release, and uptake of EVs in CC, underscoring their potential as diagnostic and prognostic biomarkers. The review also explores therapeutic strategies targeting EVs, which include modifying their release and content, utilizing them for drug delivery, genetically altering their contents, and inhibiting key cachexia pathways. Understanding the role of EVs in CC opens new avenues for diagnostic and therapeutic approaches, potentially mitigating the syndrome's impact on patient survival and quality of life.
Collapse
Affiliation(s)
- Yifeng Wang
- Department of Thoracic Surgery, Affiliated Hospital 2 of Nantong University, Nantong First People's Hospital, Nantong, 226001, P.R. China
- School of Medicine, Nantong University, Nantong, 226001, P.R. China
| | - Shengguang Ding
- Department of Thoracic Surgery, Affiliated Hospital 2 of Nantong University, Nantong First People's Hospital, Nantong, 226001, P.R. China.
| |
Collapse
|
47
|
Liao W, Lu Z, Wang C, Zhu X, Yang Y, Zhou Y, Gong P. Application and advances of biomimetic membrane materials in central nervous system disorders. J Nanobiotechnology 2024; 22:280. [PMID: 38783302 PMCID: PMC11112845 DOI: 10.1186/s12951-024-02548-8] [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: 03/08/2024] [Accepted: 05/13/2024] [Indexed: 05/25/2024] Open
Abstract
Central nervous system (CNS) diseases encompass spinal cord injuries, brain tumors, neurodegenerative diseases, and ischemic strokes. Recently, there has been a growing global recognition of CNS disorders as a leading cause of disability and death in humans and the second most common cause of death worldwide. The global burdens and treatment challenges posed by CNS disorders are particularly significant in the context of a rapidly expanding global population and aging demographics. The blood-brain barrier (BBB) presents a challenge for effective drug delivery in CNS disorders, as conventional drugs often have limited penetration into the brain. Advances in biomimetic membrane nanomaterials technology have shown promise in enhancing drug delivery for various CNS disorders, leveraging properties such as natural biological surfaces, high biocompatibility and biosafety. This review discusses recent developments in biomimetic membrane materials, summarizes the types and preparation methods of these materials, analyzes their applications in treating CNS injuries, and provides insights into the future prospects and limitations of biomimetic membrane materials.
Collapse
Affiliation(s)
- Weiquan Liao
- Department of Neurosurgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, Jiangsu, 226001, China
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu, 226001, China
| | - Zhichao Lu
- Department of Neurosurgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, Jiangsu, 226001, China
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu, 226001, China
| | - Chenxing Wang
- Department of Neurosurgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, Jiangsu, 226001, China
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu, 226001, China
| | - Xingjia Zhu
- Department of Neurosurgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, Jiangsu, 226001, China
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu, 226001, China
| | - Yang Yang
- Department of Trauma Center, Affiliated Hospital of Nantong University, Medical school of Nantong University, Nantong, Jiangsu, 226001, China
| | - Youlang Zhou
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu, 226001, China.
| | - Peipei Gong
- Department of Neurosurgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, Jiangsu, 226001, China.
- Jiangsu Medical Innovation Center, Neurological Disease Diagnosis and Treatment Center, Affiliated Hospital of Nantong University, Nantong, Jiangsu, 226001, China.
| |
Collapse
|
48
|
Qin Q, Shan Z, Xing L, Jiang Y, Li M, Fan L, Zeng X, Ma X, Zheng D, Wang H, Wang H, Liu H, Liang S, Wu L, Liang S. Synergistic effect of mesenchymal stem cell-derived extracellular vesicle and miR-137 alleviates autism-like behaviors by modulating the NF-κB pathway. J Transl Med 2024; 22:446. [PMID: 38741170 PMCID: PMC11089771 DOI: 10.1186/s12967-024-05257-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 04/29/2024] [Indexed: 05/16/2024] Open
Abstract
Autism spectrum disorder (ASD) is a multifaceted neurodevelopmental disorder predominant in childhood. Despite existing treatments, the benefits are still limited. This study explored the effectiveness of mesenchymal stem cell-derived extracellular vesicles (MSC-EVs) loaded with miR-137 in enhancing autism-like behaviors and mitigating neuroinflammation. Utilizing BTBR mice as an autism model, the study demonstrated that intranasal administration of MSC-miR137-EVs ameliorates autism-like behaviors and inhibits pro-inflammatory factors via the TLR4/NF-κB pathway. In vitro evaluation of LPS-activated BV2 cells revealed that MSC-miR137-EVs target the TLR4/NF-κB pathway through miR-137 inhibits proinflammatory M1 microglia. Moreover, bioinformatics analysis identified that MSC-EVs are rich in miR-146a-5p, which targets the TRAF6/NF-κB signaling pathway. In summary, the findings suggest that the integration of MSC-EVs with miR-137 may be a promising therapeutic strategy for ASD, which is worthy of clinical adoption.
Collapse
Affiliation(s)
- Qian Qin
- Department of Children's and Adolescent Health, Public Health College, Harbin Medical University, Harbin, 150081, China
| | - Zhiyan Shan
- Department of Histology and Embryology, Harbin Medical University, Harbin, 150081, China
| | - Lei Xing
- Department of Children's and Adolescent Health, Public Health College, Harbin Medical University, Harbin, 150081, China
| | - Yutong Jiang
- Department of Children's and Adolescent Health, Public Health College, Harbin Medical University, Harbin, 150081, China
| | - Mengyue Li
- Department of Children's and Adolescent Health, Public Health College, Harbin Medical University, Harbin, 150081, China
| | - Linlin Fan
- Department of Children's and Adolescent Health, Public Health College, Harbin Medical University, Harbin, 150081, China
| | - Xin Zeng
- Department of Children's and Adolescent Health, Public Health College, Harbin Medical University, Harbin, 150081, China
| | - Xinrui Ma
- Department of Children's and Adolescent Health, Public Health College, Harbin Medical University, Harbin, 150081, China
| | - Danyang Zheng
- Department of Children's and Adolescent Health, Public Health College, Harbin Medical University, Harbin, 150081, China
| | - Han Wang
- Department of Children's and Adolescent Health, Public Health College, Harbin Medical University, Harbin, 150081, China
| | - Hui Wang
- Department of Children's and Adolescent Health, Public Health College, Harbin Medical University, Harbin, 150081, China
| | - Hao Liu
- Department of Children's and Adolescent Health, Public Health College, Harbin Medical University, Harbin, 150081, China
| | - Shengjun Liang
- Department of Children's and Adolescent Health, Public Health College, Harbin Medical University, Harbin, 150081, China
| | - Lijie Wu
- Department of Children's and Adolescent Health, Public Health College, Harbin Medical University, Harbin, 150081, China.
| | - Shuang Liang
- Department of Children's and Adolescent Health, Public Health College, Harbin Medical University, Harbin, 150081, China.
| |
Collapse
|
49
|
Martins B, Pires M, Ambrósio AF, Girão H, Fernandes R. Contribution of extracellular vesicles for the pathogenesis of retinal diseases: shedding light on blood-retinal barrier dysfunction. J Biomed Sci 2024; 31:48. [PMID: 38730462 PMCID: PMC11088087 DOI: 10.1186/s12929-024-01036-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 04/30/2024] [Indexed: 05/12/2024] Open
Abstract
Retinal degenerative diseases, including diabetic retinopathy (DR) and age-related macular degeneration (AMD), loom as threats to vision, causing detrimental effects on the structure and function of the retina. Central to understanding these diseases, is the compromised state of the blood-retinal barrier (BRB), an effective barrier that regulates the influx of immune and inflammatory components. Whether BRB breakdown initiates retinal distress, or is a consequence of disease progression, remains enigmatic. Nevertheless, it is an indication of retinal dysfunction and potential vision loss.The intricate intercellular dialogues among retinal cell populations remain unintelligible in the complex retinal milieu, under conditions of inflammation and oxidative stress. The retina, a specialized neural tissue, sustains a ceaseless demand for oxygen and nutrients from two vascular networks. The BRB orchestrates the exchange of molecules and fluids within this specialized region, comprising the inner BRB (iBRB) and the outer BRB (oBRB). Extracellular vesicles (EVs) are small membranous structures, and act as messengers facilitating intercellular communication in this milieu.EVs, both from retinal and peripheral immune cells, increase complexity to BRB dysfunction in DR and AMD. Laden with bioactive cargoes, these EVs can modulate the retinal microenvironment, influencing disease progression. Our review delves into the multifaceted role of EVs in retinal degenerative diseases, elucidating the molecular crosstalk they orchestrate, and their microRNA (miRNA) content. By shedding light on these nanoscale messengers, from their biogenesis, release, to interaction and uptake by target cells, we aim to deepen the comprehension of BRB dysfunction and explore their therapeutic potential, therefore increasing our understanding of DR and AMD pathophysiology.
Collapse
Affiliation(s)
- Beatriz Martins
- University Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, Coimbra, 3000- 548, Portugal
- University of Coimbra, Institute of Pharmacology and Experimental Therapeutics, Faculty of Medicine, Coimbra, 3000-548, Portugal
- University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, 3004-531, Portugal
| | - Maria Pires
- University Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, Coimbra, 3000- 548, Portugal
- University of Coimbra, Institute of Pharmacology and Experimental Therapeutics, Faculty of Medicine, Coimbra, 3000-548, Portugal
- University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, 3004-531, Portugal
| | - António Francisco Ambrósio
- University Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, Coimbra, 3000- 548, Portugal
- University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, 3004-531, Portugal
- Clinical Academic Center of Coimbra (CACC), Coimbra, 3004-561, Portugal
- Association for Innovation and Biomedical Research on Light and Image (AIBILI), Coimbra, 3000-548, Portugal
| | - Henrique Girão
- University Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, Coimbra, 3000- 548, Portugal
- University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, 3004-531, Portugal
- Clinical Academic Center of Coimbra (CACC), Coimbra, 3004-561, Portugal
| | - Rosa Fernandes
- University Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, Coimbra, 3000- 548, Portugal.
- University of Coimbra, Institute of Pharmacology and Experimental Therapeutics, Faculty of Medicine, Coimbra, 3000-548, Portugal.
- University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, 3004-531, Portugal.
- Clinical Academic Center of Coimbra (CACC), Coimbra, 3004-561, Portugal.
- Association for Innovation and Biomedical Research on Light and Image (AIBILI), Coimbra, 3000-548, Portugal.
| |
Collapse
|
50
|
Moore KA, Petersen AP, Zierden HC. Microorganism-derived extracellular vesicles: emerging contributors to female reproductive health. NANOSCALE 2024; 16:8216-8235. [PMID: 38572613 DOI: 10.1039/d3nr05524h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/05/2024]
Abstract
Extracellular vesicles (EVs) are cell-derived nanoparticles that carry small molecules, nucleic acids, and proteins long distances in the body facilitating cell-cell communication. Microorganism-derived EVs mediate communication between parent cells and host cells, with recent evidence supporting their role in biofilm formation, horizontal gene transfer, and suppression of the host immune system. As lipid-bound bacterial byproducts, EVs demonstrate improved cellular uptake and distribution in vivo compared to cell-free nucleic acids, proteins, or small molecules, allowing these biological nanoparticles to recapitulate the effects of parent cells and contribute to a range of human health outcomes. Here, we focus on how EVs derived from vaginal microorganisms contribute to gynecologic and obstetric outcomes. As the composition of the vaginal microbiome significantly impacts women's health, we discuss bacterial EVs from both healthy and dysbiotic vaginal microbiota. We also examine recent work done to evaluate the role of EVs from common vaginal bacterial, fungal, and parasitic pathogens in pathogenesis of female reproductive tract disease. We highlight evidence for the role of EVs in women's health, gaps in current knowledge, and opportunities for future work. Finally, we discuss how leveraging the innate interactions between microorganisms and mammalian cells may establish EVs as a novel therapeutic modality for gynecologic and obstetric indications.
Collapse
Affiliation(s)
- Kaitlyn A Moore
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, 20742, USA.
| | - Alyssa P Petersen
- Department of Chemical & Biomolecular Engineering, University of Maryland, College Park, MD, 20742, USA
| | - Hannah C Zierden
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, 20742, USA.
- Department of Chemical & Biomolecular Engineering, University of Maryland, College Park, MD, 20742, USA
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| |
Collapse
|