51
|
Martin S, McConnell R, Harrison R, Jang SC, Sia CL, Kamerkar S, Duboff A, Jacob L, Finn J, Estes S. Therapeutic extracellular vesicle production is substantially increased by inhibition of cellular cholesterol biosynthesis. Biotechnol Bioeng 2023; 120:2685-2699. [PMID: 37060550 DOI: 10.1002/bit.28401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 03/22/2023] [Accepted: 04/01/2023] [Indexed: 04/16/2023]
Abstract
Extracellular vesicles (EVs) are a new therapeutic modality with the promise to treat many diseases through their ability to deliver diverse molecular cargo. As with other emerging modalities transitioning into the industrialization phase, all aspects of the manufacturing process are rich with opportunities to enhance the ability to deliver these medicines to patients. With the goal of improving cell culture EV productivity, we have utilized high throughput siRNA screens to identify the underlying genetic pathways that regulate EV productivity to inform rational host cell line engineering and media development approaches. The screens identified multiple metabolic pathways of potential interest; one of which was validated and shown to be a ready implementable, cost-effective strategy to increase EV titers. We show that both EV volumetric and specific productivity from HEK293 and CHO-S were increased in a dose and cell line-dependent manner up to ninefold when cholesterol synthesis was inhibited by the inclusion of statins in the cell culture media. In addition, we show in response to statin treatment, elevation of EV markers in mesenchymal stem cell (MSC) cell culture media suggesting this approach can also be applicable to MSC EVs. Furthermore, we show that the EVs produced from statin-treated HEK293 cultures are effectively loaded by both endogenous and exogenous loading methods and have equivalent in vitro or in vivo potency relative to EVs from untreated cultures.
Collapse
Affiliation(s)
| | | | | | - Su Chul Jang
- Codiak BioSciences, Cambridge, Massachusetts, USA
| | | | | | - Anna Duboff
- Codiak BioSciences, Cambridge, Massachusetts, USA
| | - Lisa Jacob
- Codiak BioSciences, Cambridge, Massachusetts, USA
| | | | - Scott Estes
- Codiak BioSciences, Cambridge, Massachusetts, USA
| |
Collapse
|
52
|
Zou Z, Li H, Xu G, Hu Y, Zhang W, Tian K. Current Knowledge and Future Perspectives of Exosomes as Nanocarriers in Diagnosis and Treatment of Diseases. Int J Nanomedicine 2023; 18:4751-4778. [PMID: 37635911 PMCID: PMC10454833 DOI: 10.2147/ijn.s417422] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 07/29/2023] [Indexed: 08/29/2023] Open
Abstract
Exosomes, as natural nanocarriers, characterized with low immunogenicity, non-cytotoxicity and targeted delivery capability, which have advantages over synthetic nanocarriers. Recently, exosomes have shown great potential as diagnostic markers for diseases and are also considered as a promising cell-free therapy. Engineered exosomes have significantly enhanced the efficacy and precision of delivering therapeutic agents, and are currently being extensively employed in targeted therapeutic investigations for various ailments, including oncology, inflammatory disorders, and degenerative conditions. Particularly, engineered exosomes enable therapeutic agent loading, targeted modification, evasion of MPS phagocytosis, intelligent control, and bioimaging, and have been developed as multifunctional nano-delivery platforms in recent years. The utilization of bioactive scaffolds that are loaded with exosome delivery has been shown to substantially augment retention, extend exosome release, and enhance efficacy. This approach has advanced from conventional hydrogels to nanocomposite hydrogels, nanofiber hydrogels, and 3D printing, resulting in superior physical and biological properties that effectively address the limitations of natural scaffolds. Additionally, plant-derived exosomes, which can participate in gut flora remodeling via oral administration, are considered as an ideal delivery platform for the treatment of intestinal diseases. Consequently, there is great interest in exosomes and exosomes as nanocarriers for therapeutic and diagnostic applications. This comprehensive review provides an overview of the biogenesis, composition, and isolation methods of exosomes. Additionally, it examines the pathological and diagnostic mechanisms of exosomes in various diseases, including tumors, degenerative disorders, and inflammatory conditions. Furthermore, this review highlights the significance of gut microbial-derived exosomes. Strategies and specific applications of engineered exosomes and bioactive scaffold-loaded exosome delivery are further summarized, especially some new techniques such as large-scale loading technique, macromolecular loading technique, development of multifunctional nano-delivery platforms and nano-scaffold-loaded exosome delivery. The potential benefits of using plant-derived exosomes for the treatment of gut-related diseases are also discussed. Additionally, the challenges, opportunities, and prospects of exosome-based nanocarriers for disease diagnosis and treatment are summarized from both preclinical and clinical viewpoints.
Collapse
Affiliation(s)
- Zaijun Zou
- Department of Sports Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, 116011, People’s Republic of China
- School of Graduates, Dalian Medical University, Dalian, Liaoning, 116000, People’s Republic of China
| | - Han Li
- Department of Sports Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, 116011, People’s Republic of China
- School of Graduates, Dalian Medical University, Dalian, Liaoning, 116000, People’s Republic of China
| | - Gang Xu
- Department of Sports Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, 116011, People’s Republic of China
- Key Laboratory of Molecular Mechanism for Repair and Remodeling of Orthopaedic Disease, Dalian, Liaoning Province, 116011, People’s Republic of China
| | - Yunxiang Hu
- School of Graduates, Dalian Medical University, Dalian, Liaoning, 116000, People’s Republic of China
| | - Weiguo Zhang
- Department of Sports Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, 116011, People’s Republic of China
- Key Laboratory of Molecular Mechanism for Repair and Remodeling of Orthopaedic Disease, Dalian, Liaoning Province, 116011, People’s Republic of China
| | - Kang Tian
- Department of Sports Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, 116011, People’s Republic of China
- Key Laboratory of Molecular Mechanism for Repair and Remodeling of Orthopaedic Disease, Dalian, Liaoning Province, 116011, People’s Republic of China
| |
Collapse
|
53
|
Bao C, Xiang H, Chen Q, Zhao Y, Gao Q, Huang F, Mao L. A Review of Labeling Approaches Used in Small Extracellular Vesicles Tracing and Imaging. Int J Nanomedicine 2023; 18:4567-4588. [PMID: 37588627 PMCID: PMC10426735 DOI: 10.2147/ijn.s416131] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 07/26/2023] [Indexed: 08/18/2023] Open
Abstract
Small extracellular vesicles (sEVs), a subset of extracellular vesicles (EVs) originating from the endosomal compartment, are a kind of lipid bilayer vesicles released by almost all types of cells, serving as natural carriers of nucleic acids, proteins, and lipids for intercellular communication and transfer of bioactive molecules. The current findings suggest their vital role in physiological and pathological processes. Various sEVs labeling techniques have been developed for the more advanced study of the function, mode of action, bio-distribution, and related information of sEVs. In this review, we summarize the existing and emerging sEVs labeling techniques, including fluorescent labeling, radioisotope labeling, nanoparticle labeling, chemical contrast agents labeling, and label-free technique. These approaches will pave the way for an in-depth study of sEVs. We present a systematic and comprehensive review of the principles, advantages, disadvantages, and applications of these techniques, to help promote applications of these labeling approaches in future research on sEVs.
Collapse
Affiliation(s)
- Chenxuan Bao
- Department of Laboratory Medicine, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, Jiangsu, People’s Republic of China
| | - Huayuan Xiang
- Department of Laboratory Medicine, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, Jiangsu, People’s Republic of China
| | - Qiaoqiao Chen
- Department of Laboratory Medicine, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, Jiangsu, People’s Republic of China
- Department of Laboratory Medicine, the Affiliated People’s Hospital, Jiangsu University, Zhenjiang, Jiangsu, People’s Republic of China
| | - Yuxue Zhao
- Department of Laboratory Medicine, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, Jiangsu, People’s Republic of China
- Department of Laboratory Medicine, the Affiliated People’s Hospital, Jiangsu University, Zhenjiang, Jiangsu, People’s Republic of China
| | - Qianqian Gao
- Department of Laboratory Medicine, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, Jiangsu, People’s Republic of China
| | - Feng Huang
- Department of Laboratory Medicine, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, Jiangsu, People’s Republic of China
| | - Lingxiang Mao
- Department of Laboratory Medicine, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, Jiangsu, People’s Republic of China
- Department of Laboratory Medicine, the Affiliated People’s Hospital, Jiangsu University, Zhenjiang, Jiangsu, People’s Republic of China
| |
Collapse
|
54
|
Zheng W, Rädler J, Sork H, Niu Z, Roudi S, Bost JP, Görgens A, Zhao Y, Mamand DR, Liang X, Wiklander OPB, Lehto T, Gupta D, Nordin JZ, El Andaloussi S. Identification of scaffold proteins for improved endogenous engineering of extracellular vesicles. Nat Commun 2023; 14:4734. [PMID: 37550290 PMCID: PMC10406850 DOI: 10.1038/s41467-023-40453-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 07/27/2023] [Indexed: 08/09/2023] Open
Abstract
Extracellular vesicles (EVs) are gaining ground as next-generation drug delivery modalities. Genetic fusion of the protein of interest to a scaffold protein with high EV-sorting ability represents a robust cargo loading strategy. To address the paucity of such scaffold proteins, we leverage a simple and reliable assay that can distinguish intravesicular cargo proteins from surface- as well as non-vesicular proteins and compare the EV-sorting potential of 244 candidate proteins. We identify 24 proteins with conserved EV-sorting abilities across five types of producer cells. TSPAN2 and TSPAN3 emerge as lead candidates and outperform the well-studied CD63 scaffold. Importantly, these engineered EVs show promise as delivery vehicles in cell cultures and mice as demonstrated by efficient transfer of luminal cargo proteins as well as surface display of different functional entities. The discovery of these scaffolds provides a platform for EV-based engineering.
Collapse
Affiliation(s)
- Wenyi Zheng
- Division of Biomolecular and Cellular Medicine, Department of Laboratory Medicine, Karolinska Institutet, Huddinge, Sweden
| | - Julia Rädler
- Division of Biomolecular and Cellular Medicine, Department of Laboratory Medicine, Karolinska Institutet, Huddinge, Sweden
| | - Helena Sork
- Institute of Technology, University of Tartu, Tartu, Estonia
| | - Zheyu Niu
- Division of Biomolecular and Cellular Medicine, Department of Laboratory Medicine, Karolinska Institutet, Huddinge, Sweden
- Department of Hepatobiliary Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Samantha Roudi
- Division of Biomolecular and Cellular Medicine, Department of Laboratory Medicine, Karolinska Institutet, Huddinge, Sweden
| | - Jeremy P Bost
- Division of Biomolecular and Cellular Medicine, Department of Laboratory Medicine, Karolinska Institutet, Huddinge, Sweden
| | - André Görgens
- Division of Biomolecular and Cellular Medicine, Department of Laboratory Medicine, Karolinska Institutet, Huddinge, Sweden
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Ying Zhao
- Division of Biomolecular and Cellular Medicine, Department of Laboratory Medicine, Karolinska Institutet, Huddinge, Sweden
- Clinical Research Center, Karolinska University Hospital, Stockholm, Sweden
| | - Doste R Mamand
- Division of Biomolecular and Cellular Medicine, Department of Laboratory Medicine, Karolinska Institutet, Huddinge, Sweden
| | - Xiuming Liang
- Division of Biomolecular and Cellular Medicine, Department of Laboratory Medicine, Karolinska Institutet, Huddinge, Sweden
| | - Oscar P B Wiklander
- Division of Biomolecular and Cellular Medicine, Department of Laboratory Medicine, Karolinska Institutet, Huddinge, Sweden
| | - Taavi Lehto
- Division of Biomolecular and Cellular Medicine, Department of Laboratory Medicine, Karolinska Institutet, Huddinge, Sweden
- Institute of Technology, University of Tartu, Tartu, Estonia
| | - Dhanu Gupta
- Division of Biomolecular and Cellular Medicine, Department of Laboratory Medicine, Karolinska Institutet, Huddinge, Sweden
| | - Joel Z Nordin
- Division of Biomolecular and Cellular Medicine, Department of Laboratory Medicine, Karolinska Institutet, Huddinge, Sweden
| | - Samir El Andaloussi
- Division of Biomolecular and Cellular Medicine, Department of Laboratory Medicine, Karolinska Institutet, Huddinge, Sweden.
| |
Collapse
|
55
|
Luo S, Chen J, Xu F, Chen H, Li Y, Li W. Dendritic Cell-Derived Exosomes in Cancer Immunotherapy. Pharmaceutics 2023; 15:2070. [PMID: 37631284 PMCID: PMC10457773 DOI: 10.3390/pharmaceutics15082070] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 07/21/2023] [Accepted: 07/25/2023] [Indexed: 08/27/2023] Open
Abstract
Exosomes are nanoscale vesicles released by diverse types of cells for complex intercellular communication. Numerous studies have shown that exosomes can regulate the body's immune response to tumor cells and interfere with the tumor microenvironment (TME). In clinical trials on dendritic cell (DC)-based antitumor vaccines, no satisfactory results have been achieved. However, recent studies suggested that DC-derived exosomes (DEXs) may be superior to DC-based antitumor vaccines in avoiding tumor cell-mediated immunosuppression. DEXs contain multiple DC-derived surface markers that capture tumor-associated antigens (TAAs) and promote immune cell-dependent tumor rejection. These findings indicate the necessity of the further development and improvement of DEX-based cell-free vaccines to complement chemotherapy, radiotherapy, and other immunotherapies. In this review, we highlighted the recent progress of DEXs in cancer immunotherapy, particularly by concentrating on landmark studies and the biological characterization of DEXs, and we summarized their important role in the tumor immune microenvironment (TIME) and clinical application in targeted cancer immunotherapy. This review could enhance comprehension of advances in cancer immunotherapy and contribute to the elucidation of how DEXs regulate the TIME, thereby providing a reference for utilizing DEX-based vaccines in clinical practice.
Collapse
Affiliation(s)
- Shumin Luo
- Beijing Institute of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing 100069, China; (S.L.); (J.C.); (F.X.); (Y.L.)
| | - Jing Chen
- Beijing Institute of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing 100069, China; (S.L.); (J.C.); (F.X.); (Y.L.)
| | - Fang Xu
- Beijing Institute of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing 100069, China; (S.L.); (J.C.); (F.X.); (Y.L.)
| | - Huan Chen
- Integrated Chinese and Western Medicine Center, Beijing Youan Hospital, Capital Medical University, Beijing 100069, China;
| | - Yiru Li
- Beijing Institute of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing 100069, China; (S.L.); (J.C.); (F.X.); (Y.L.)
| | - Weihua Li
- Beijing Institute of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing 100069, China; (S.L.); (J.C.); (F.X.); (Y.L.)
| |
Collapse
|
56
|
Maalouf KE, Vaine CA, Frederick DM, Yoshinaga A, Obuchi W, Mahjoum S, Nieland L, Al Ali J, Bragg DC, Breakefield XO, Breyne K. Tracking human neurologic disease status in mouse brain/plasma using reporter-tagged, EV-associated biomarkers. Mol Ther 2023; 31:2206-2219. [PMID: 37198883 PMCID: PMC10362415 DOI: 10.1016/j.ymthe.2023.05.011] [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: 11/02/2022] [Revised: 04/17/2023] [Accepted: 05/12/2023] [Indexed: 05/19/2023] Open
Abstract
X-linked dystonia-parkinsonism (XDP) is a neurodegenerative disease caused by a retrotransposon insertion in intron 32 of the TAF1 gene. This insertion causes mis-splicing of intron 32 (TAF1-32i) and reduced TAF1 levels. TAF1-32i transcript is unique to XDP patient cells and can be detected in their extracellular vesicles (EVs). We engrafted patient and control iPSC-derived neural progenitor cells (hNPCs) into the striatum of mice. To track TAF1-32i transcript spread by EVs, we transduced the brain-implanted hNPCs with a lentiviral construct called ENoMi, which consists of a re-engineered tetraspanin scaffold tagged with bioluminescent and fluorescent reporter proteins under an EF-1α promoter. Alongside this improved detection in ENoMi-hNPCs-derived EVs, their surface allows specific immunocapture purification, thereby facilitating TAF1-32i analysis. Using this ENoMi-labeling method, TAF1-32i was demonstrated in EVs released from XDP hNPCs implanted in mouse brains. Post-implantation of ENoMi-XDP hNPCs, TAF1-32i transcript was retrieved in EVs isolated from mouse brain and blood, and levels increased over time in plasma. We compared and combined our EV isolation technique to analyze XDP-derived TAF1-32i with other techniques, including size exclusion chromatography and Exodisc. Overall, our study demonstrates the successful engraftment of XDP patient-derived hNPCs in mice as a tool for monitoring disease markers with EVs.
Collapse
Affiliation(s)
- Katia E Maalouf
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; The Collaborative Center for X-linked Dystonia-Parkinsonism, Massachusetts General Hospital, Charlestown, MA 02129, USA
| | - Christine A Vaine
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; The Collaborative Center for X-linked Dystonia-Parkinsonism, Massachusetts General Hospital, Charlestown, MA 02129, USA
| | - Dawn M Frederick
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Akiko Yoshinaga
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Wataru Obuchi
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Shadi Mahjoum
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Lisa Nieland
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Jamal Al Ali
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - D Cristopher Bragg
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; The Collaborative Center for X-linked Dystonia-Parkinsonism, Massachusetts General Hospital, Charlestown, MA 02129, USA
| | - Xandra O Breakefield
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; The Collaborative Center for X-linked Dystonia-Parkinsonism, Massachusetts General Hospital, Charlestown, MA 02129, USA; Center for Molecular Imaging Research, Department of Radiology, Massachusetts General Hospital and Program in Neuroscience, Harvard Medical School, Boston, MA 02114, USA.
| | - Koen Breyne
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; Center for Molecular Imaging Research, Department of Radiology, Massachusetts General Hospital and Program in Neuroscience, Harvard Medical School, Boston, MA 02114, USA.
| |
Collapse
|
57
|
Yamamoto A, Toba M, Takahashi Y, Takakura Y. Pharmacokinetic Approach for the Elucidation of Elevated Plasma Small Extracellular Vesicle (sEV) Concentration in Cancer. J Pharm Sci 2023; 112:1967-1974. [PMID: 37001861 DOI: 10.1016/j.xphs.2023.03.017] [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: 01/07/2023] [Revised: 03/24/2023] [Accepted: 03/24/2023] [Indexed: 04/03/2023]
Abstract
The abundance of circulating plasma small extracellular vesicles (sEVs) has been reported to be elevated in cancer; however, the underlying mechanism remains unclear. In this study, a pharmacokinetic approach was used to determine the factors contributing to elevated plasma sEV levels during cancer in a tumor-bearing mouse model. Mouse plasma-derived sEVs (MP-sEVs) isolated from tumor-bearing mice showed increased protein concentrations and physicochemical characteristics comparable to MP-sEVs isolated from healthy mice. The steady-state concentration of sEVs is determined by the balance between the MP-sEV production and clearance. Thus, to determine whether tumorigenesis influences sEV clearance, isolated MP-sEVs were intravenously administered to either tumor-bearing or healthy mice. The results showed minimal differences in sEV clearance rates, suggesting that sEV production is the driving force of elevated MP-sEV concentrations. Lastly, CD63-gLuc stably expressing B16BL6-bearing mice were used to estimate the contribution of tumor cell-derived sEVs in the plasma. The gLuc activity of the MP-sEVs isolated was below the limit of detection, and it was estimated that the tumor cell-derived sEVs comprised at most 0.5% of the total MP-sEVs. Taken together, these results suggest that cells other than tumor cells contribute to elevated plasma sEV levels in cancer.
Collapse
Affiliation(s)
- Aki Yamamoto
- Department of Biopharmaceutics and Drug Metabolism, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| | - Mihiro Toba
- Department of Biopharmaceutics and Drug Metabolism, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| | - Yuki Takahashi
- Department of Biopharmaceutics and Drug Metabolism, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| | - Yoshinobu Takakura
- Department of Biopharmaceutics and Drug Metabolism, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| |
Collapse
|
58
|
Petroni D, Fabbri C, Babboni S, Menichetti L, Basta G, Del Turco S. Extracellular Vesicles and Intercellular Communication: Challenges for In Vivo Molecular Imaging and Tracking. Pharmaceutics 2023; 15:1639. [PMID: 37376087 PMCID: PMC10301899 DOI: 10.3390/pharmaceutics15061639] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 05/23/2023] [Accepted: 05/30/2023] [Indexed: 06/29/2023] Open
Abstract
Extracellular vesicles (EVs) are a heterogeneous class of cell-derived membrane vesicles released by various cell types that serve as mediators of intercellular signaling. When released into circulation, EVs may convey their cargo and serve as intermediaries for intracellular communication, reaching nearby cells and possibly also distant organs. In cardiovascular biology, EVs released by activated or apoptotic endothelial cells (EC-EVs) disseminate biological information at short and long distances, contributing to the development and progression of cardiovascular disease and related disorders. The significance of EC-EVs as mediators of cell-cell communication has advanced, but a thorough knowledge of the role that intercommunication plays in healthy and vascular disease is still lacking. Most data on EVs derive from in vitro studies, but there are still little reliable data available on biodistribution and specific homing EVs in vivo tissues. Molecular imaging techniques for EVs are crucial to monitoring in vivo biodistribution and the homing of EVs and their communication networks both in basal and pathological circumstances. This narrative review provides an overview of EC-EVs, trying to highlight their role as messengers of cell-cell interaction in vascular homeostasis and disease, and describes emerging applications of various imaging modalities for EVs visualization in vivo.
Collapse
Affiliation(s)
- Debora Petroni
- Institute of Clinical Physiology, CNR San Cataldo Research Area, Via Moruzzi 1, 56124 Pisa, Italy
| | - Costanza Fabbri
- Institute of Clinical Physiology, CNR San Cataldo Research Area, Via Moruzzi 1, 56124 Pisa, Italy
- Institute of Life Sciences, Scuola Superiore Sant’Anna, 56127 Pisa, Italy
| | - Serena Babboni
- Institute of Clinical Physiology, CNR San Cataldo Research Area, Via Moruzzi 1, 56124 Pisa, Italy
| | - Luca Menichetti
- Institute of Clinical Physiology, CNR San Cataldo Research Area, Via Moruzzi 1, 56124 Pisa, Italy
| | - Giuseppina Basta
- Institute of Clinical Physiology, CNR San Cataldo Research Area, Via Moruzzi 1, 56124 Pisa, Italy
| | - Serena Del Turco
- Institute of Clinical Physiology, CNR San Cataldo Research Area, Via Moruzzi 1, 56124 Pisa, Italy
| |
Collapse
|
59
|
Cober ND, Rowe K, Deng Y, Benavente‐Babace A, Courtman DW, Godin M, Stewart DJ. Targeting extracellular vesicle delivery to the lungs by microgel encapsulation. JOURNAL OF EXTRACELLULAR BIOLOGY 2023; 2:e94. [PMID: 38938918 PMCID: PMC11080904 DOI: 10.1002/jex2.94] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 04/29/2023] [Accepted: 05/17/2023] [Indexed: 06/29/2024]
Abstract
Extracellular vesicles (EVs) secreted by stem and progenitor cells have significant potential as cell-free 'cellular' therapeutics. Yet, small EVs (<200 nm) are rapidly cleared after systemic administration, mainly by the liver, presenting challenges targeting EVs to a specific organ or tissue. Microencapsulation using natural nano-porous hydrogels (microgels) has been shown to enhance engraftment and increase the survival of transplanted cells. We sought to encapsulate EVs within microgels to target their delivery to the lung by virtue of their size-based retention within the pulmonary microcirculation. Mesenchymal stromal cell (MSC) derived EVs were labelled with the lipophilic dye (DiR) and encapsulated within agarose-gelatin microgels. Endothelial cells and bone marrow derived macrophages were able to take up EVs encapsulated in microgels in vitro, but less efficiently than the uptake of free EVs. Following intrajugular administration, microgel encapsulated EVs were selectively retained within the lungs for 72h, while free EVs were rapidly cleared by the liver. Furthermore, microgel-loaded EVs demonstrated greater uptake by lung cells, in particular CD45+ immune cells, as assessed by flow cytometry compared to free EVs. Microencapsulation of EVs may be a novel tool for enhancing the targeted delivery of EVs for future therapeutic applications.
Collapse
Affiliation(s)
- Nicholas D. Cober
- Sinclair Centre for Regenerative MedicineOttawa Hospital Research InstituteOttawaOntarioCanada
- Faculty of MedicineDepartment of Cellular and Molecular MedicineUniversity of OttawaOttawaOntarioCanada
| | - Katelynn Rowe
- Sinclair Centre for Regenerative MedicineOttawa Hospital Research InstituteOttawaOntarioCanada
| | - Yupu Deng
- Sinclair Centre for Regenerative MedicineOttawa Hospital Research InstituteOttawaOntarioCanada
| | | | - David W. Courtman
- Sinclair Centre for Regenerative MedicineOttawa Hospital Research InstituteOttawaOntarioCanada
| | - Michel Godin
- Faculty of Science, Department of PhysicsUniversity of OttawaOttawaOntarioCanada
| | - Duncan J. Stewart
- Sinclair Centre for Regenerative MedicineOttawa Hospital Research InstituteOttawaOntarioCanada
- Faculty of MedicineDepartment of Cellular and Molecular MedicineUniversity of OttawaOttawaOntarioCanada
| |
Collapse
|
60
|
Ashique S, Anand K. Radiolabelled Extracellular Vesicles as Imaging Modalities for Precise Targeted Drug Delivery. Pharmaceutics 2023; 15:pharmaceutics15051426. [PMID: 37242668 DOI: 10.3390/pharmaceutics15051426] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 04/20/2023] [Accepted: 04/26/2023] [Indexed: 05/28/2023] Open
Abstract
Extracellular vesicles (ECVs) have been abandoned as bio-inspired drug delivery systems (DDS) in the biomedical field. ECVs have a natural ability to cross over extracellular and intracellular barriers, making them superior to manufactured nanoparticles. Additionally, they have the ability to move beneficial biomolecules among far-flung bodily cells. These advantages and the accomplishment of favorable in vivo results convincingly show the value of ECVs in medication delivery. The usage of ECVs is constantly being improved, as it might be difficult to develop a consistent biochemical strategy that is in line with their useful clinical therapeutic uses. Extracellular vesicles (ECVs) have the potential to enhance the therapy of diseases. Imaging technologies, particularly radiolabelled imaging, have been exploited for non-invasive tracking to better understand their in vivo activity.
Collapse
Affiliation(s)
- Sumel Ashique
- Department of Pharmaceutics, Bharat Institute of Technology (BIT), School of Pharmacy, Meerut 250103, India
| | - Krishnan Anand
- Department of Chemical Pathology, School of Pathology, Faculty of Health Sciences, University of the Free State, Bloemfontein 9300, South Africa
| |
Collapse
|
61
|
Nishi K, Izumi H, Tomonaga T, Nagano C, Morimoto Y, Horie S. IL-6-Mediated Upregulated miRNAs in Extracellular Vesicles Derived from Lund Human Mesencephalic (LUHMES) Cells: Effects on Astrocytes and Microglia. Biomolecules 2023; 13:biom13050718. [PMID: 37238588 DOI: 10.3390/biom13050718] [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: 03/06/2023] [Revised: 04/07/2023] [Accepted: 04/19/2023] [Indexed: 05/28/2023] Open
Abstract
Psychological stress plays a major role in depression, and interleukin-6 (IL-6) is elevated during depression and psychological stress. MicroRNAs (miRNAs) in extracellular vesicles (EVs), including exosomes and microvesicles, suppress mRNA expression in other cells when endocytosed. In this study, we analyzed the effect of IL-6 on EVs secreted by neural precursor cells. Cells from the human immortalized neural precursor cell line LUHMES were treated with IL-6. EVs were collected using a nanofiltration method. We then analyzed the uptake of LUHMES-derived EVs by astrocytes (ACs) and microglia (MG). Microarray analysis of miRNAs was performed using EV-incorporated RNA and intracellular RNA from ACs and MG to search for increased numbers of miRNAs. We applied the miRNAs to ACs and MG, and examined the cells for suppressed mRNAs. IL-6 increased several miRNAs in the EVs. Three of these miRNAs were originally low in ACs and MG (hsa-miR-135a-3p, hsa-miR-6790-3p, and hsa-miR-11399). In ACs and MG, hsa-miR-6790-3p and hsa-miR-11399 suppressed four mRNAs involved in nerve regeneration (NREP, KCTD12, LLPH, and CTNND1). IL-6 altered the types of miRNAs in EVs derived from neural precursor cells, by which mRNAs involved in nerve regeneration were decreased in ACs and MG. These findings provide new insights into the involvement of IL-6 in stress and depression.
Collapse
Affiliation(s)
- Kento Nishi
- Center for Stress-Related Disease Control and Prevention, University of Occupational and Environmental Health, Kitakyushu 807-8555, Japan
- Department of Health Policy and Management, Institute of Industrial Ecological Sciences, University of Occupational and Environmental Health, Kitakyushu 807-8555, Japan
| | - Hiroto Izumi
- Center for Stress-Related Disease Control and Prevention, University of Occupational and Environmental Health, Kitakyushu 807-8555, Japan
- Department of Occupational Pneumology, Institute of Industrial Ecological Sciences, University of Occupational and Environmental Health, Kitakyushu 807-8555, Japan
| | - Taisuke Tomonaga
- Department of Occupational Pneumology, Institute of Industrial Ecological Sciences, University of Occupational and Environmental Health, Kitakyushu 807-8555, Japan
| | - Chikage Nagano
- Center for Stress-Related Disease Control and Prevention, University of Occupational and Environmental Health, Kitakyushu 807-8555, Japan
- Department of Health Policy and Management, Institute of Industrial Ecological Sciences, University of Occupational and Environmental Health, Kitakyushu 807-8555, Japan
| | - Yasuo Morimoto
- Center for Stress-Related Disease Control and Prevention, University of Occupational and Environmental Health, Kitakyushu 807-8555, Japan
- Department of Occupational Pneumology, Institute of Industrial Ecological Sciences, University of Occupational and Environmental Health, Kitakyushu 807-8555, Japan
| | - Seichi Horie
- Center for Stress-Related Disease Control and Prevention, University of Occupational and Environmental Health, Kitakyushu 807-8555, Japan
- Department of Health Policy and Management, Institute of Industrial Ecological Sciences, University of Occupational and Environmental Health, Kitakyushu 807-8555, Japan
| |
Collapse
|
62
|
Gupta D, Wiklander OP, Wood MJ, El-Andaloussi S. Biodistribution of therapeutic extracellular vesicles. EXTRACELLULAR VESICLES AND CIRCULATING NUCLEIC ACIDS 2023; 4:170-190. [PMID: 39697988 PMCID: PMC11648525 DOI: 10.20517/evcna.2023.12] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 03/27/2023] [Accepted: 04/03/2023] [Indexed: 12/20/2024]
Abstract
The field of extracellular vesicles (EVs) has seen a tremendous paradigm shift in the past two decades, from being regarded as cellular waste bags to being considered essential mediators in intercellular communication. Their unique ability to transfer macromolecules across cells and biological barriers has made them a rising star in drug delivery. Mounting evidence suggests that EVs can be explored as efficient drug delivery vehicles for a range of therapeutic macromolecules. In contrast to many synthetic delivery systems, these vesicles appear exceptionally well tolerated in vivo. This tremendous development in the therapeutic application of EVs has been made through technological advancement in labelling and understanding the in vivo biodistribution of EVs. Here in this review, we have summarised the recent findings in EV in vivo pharmacokinetics and discussed various biological barriers that need to be surpassed to achieve tissue-specific delivery.
Collapse
Affiliation(s)
- Dhanu Gupta
- Department of Paediatrics. University of Oxford, Oxford OX3 7TY, UK
- Biomolecular Medicine, Division of Biomolecular and Cellular Medicine, Department of Laboratory Medicine, Karolinska Institutet, Huddinge 14151, Sweden
| | - Oscar P.B Wiklander
- Biomolecular Medicine, Division of Biomolecular and Cellular Medicine, Department of Laboratory Medicine, Karolinska Institutet, Huddinge 14151, Sweden
| | - Matthew J.A Wood
- Department of Paediatrics. University of Oxford, Oxford OX3 7TY, UK
| | - Samir El-Andaloussi
- Biomolecular Medicine, Division of Biomolecular and Cellular Medicine, Department of Laboratory Medicine, Karolinska Institutet, Huddinge 14151, Sweden
| |
Collapse
|
63
|
Therapeutic potential of extracellular vesicles in neurodegenerative disorders. HANDBOOK OF CLINICAL NEUROLOGY 2023; 193:243-266. [PMID: 36803815 DOI: 10.1016/b978-0-323-85555-6.00017-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/18/2023]
Abstract
Neurodegenerative disorders are characterized by complex multifactorial pathogeneses, thus posing a challenge for standard therapeutic approaches that tend to focus only on one underlying disease aspect. For systemically administered drugs, the blood-brain barrier (BBB) is yet another major obstacle to overcome. In this context, naturally occurring extracellular vesicles (EVs) with intrinsic ability to cross the BBB have been investigated as therapeutics for various diseases, including Alzheimer's and Parkinson's diseases. EVs are cell-derived, lipid membrane-enclosed vesicles carrying a broad spectrum of biologically active molecules, which play a crucial role in intercellular communication. In a therapeutic context, mesenchymal stem cell (MSC)-derived EVs are in the spotlight because they reflect the therapeutic properties of their parental cells and, thus, hold promise as independent cell-free therapeutics. On the other hand, EVs can be used as drug delivery vehicles by modifying their surface or content, e.g., by decorating the surface with brain-specific ligands or loading the EVs with therapeutic RNAs or proteins, thus further enhancing the EV's targeting and therapeutic potency, respectively. Although EVs have been deemed safe for use in humans, some obstacles remain that prevent their progression into clinics. This review scrutinizes the promises and challenges of EV-based treatments for neurodegenerative disorders.
Collapse
|
64
|
Biodistribution of Intratracheal, Intranasal, and Intravenous Injections of Human Mesenchymal Stromal Cell-Derived Extracellular Vesicles in a Mouse Model for Drug Delivery Studies. Pharmaceutics 2023; 15:pharmaceutics15020548. [PMID: 36839873 PMCID: PMC9964290 DOI: 10.3390/pharmaceutics15020548] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 01/31/2023] [Accepted: 02/03/2023] [Indexed: 02/11/2023] Open
Abstract
Mesenchymal stromal cell-derived extracellular vesicles (MSC-EVs) are extensively studied as therapeutic tools. Evaluation of their biodistribution is fundamental to understanding MSC-EVs' impact on target organs. In our work, MSC-EVs were initially labeled with DiR, a fluorescent lipophilic dye, and administered to BALB/c mice (2.00 × 1010 EV/mice) through the following routes: intravenous (IV), intratracheal (IT) and intranasal (IN). DiR-labeled MSC-EVs were monitored immediately after injection, and after 3 and 24 hours (h). Whole-body analysis, 3 h after IV injection, showed an accumulation of MSC-EVs in the mice abdominal region, compared to IT and IN, where EVs mainly localized at the levels of the chest and brain region, respectively. After 24 h, EV-injected mice retained a stronger positivity in the same regions identified after 3 h from injection. The analyses of isolated organs confirmed the accumulation of EVs in the spleen and liver after IV administration. Twenty-four hours after the IT injection of MSC-EVs, a stronger positivity was detected selectively in the isolated lungs, while for IN, the signal was confined to the brain. In conclusion, these results show that local administration of EVs can increase their concentration in selective organs, limiting their systemic biodistribution and possibly the extra-organ effects. Biodistribution studies can help in the selection of the most appropriate way of administration of MSC-EVs for the treatment of different diseases.
Collapse
|
65
|
Itagaki M, Nasu Y, Sugiyama C, Nakase I, Kamei N. A universal method to analyze cellular internalization mechanisms via endocytosis without non-specific cross-effects. FASEB J 2023; 37:e22764. [PMID: 36624697 DOI: 10.1096/fj.202201780r] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 12/16/2022] [Accepted: 12/27/2022] [Indexed: 01/11/2023]
Abstract
Endocytosis is an essential biological process for nutrient absorption and intercellular communication; it can also be used to accelerate the cellular internalization of drug delivery carriers. Clarifying the cellular uptake mechanisms of unidentified endogenous and exogenous molecules and designing new effective drug delivery systems require an accurate, specific endocytosis analysis methodology. Therefore, we developed a method to specifically evaluate cellular internalization via three main endocytic pathways: clathrin- and caveolae-mediated endocytosis, and macropinocytosis. We first revealed that most known endocytosis inhibitors had no specific inhibitory effect or were cytotoxic. Second, we successfully established an alternative method using small interfering RNA to knock down dynamin-2 and caveolin-1, which are necessary for clathrin- and caveolae-mediated endocytosis, in HeLa cells. Third, we established another method to specifically analyze macropinocytosis using rottlerin on A431 cells. Finally, we validated the proposed methods by testing the cellular internalization of a biological molecule (insulin) and carriers (nanoparticles and cell-penetrating peptides). Through this study, we established versatile methods to precisely and specifically evaluate endocytosis of newly developed biopharmaceuticals or drug delivery systems.
Collapse
Affiliation(s)
- Mai Itagaki
- Laboratory of Drug Delivery Systems, Graduate School of Pharmaceutical Sciences, Kobe Gakuin University, Kobe, Japan
| | - Yoshinori Nasu
- Laboratory of Drug Delivery Systems, Faculty of Pharmaceutical Sciences, Kobe Gakuin University, Kobe, Japan
| | - Chiaki Sugiyama
- Laboratory of Drug Delivery Systems, Faculty of Pharmaceutical Sciences, Kobe Gakuin University, Kobe, Japan
| | - Ikuhiko Nakase
- Graduate School of Science, Osaka Metropolitan University, Osaka, Japan
| | - Noriyasu Kamei
- Laboratory of Drug Delivery Systems, Faculty of Pharmaceutical Sciences, Kobe Gakuin University, Kobe, Japan
| |
Collapse
|
66
|
Li H, Ding Y, Huang J, Zhao Y, Chen W, Tang Q, An Y, Chen R, Hu C. Angiopep-2 Modified Exosomes Load Rifampicin with Potential for Treating Central Nervous System Tuberculosis. Int J Nanomedicine 2023; 18:489-503. [PMID: 36733407 PMCID: PMC9888470 DOI: 10.2147/ijn.s395246] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 01/18/2023] [Indexed: 01/28/2023] Open
Abstract
Background Central nervous system tuberculosis (CNS-TB) is the most devastating form of extrapulmonary tuberculosis. Rifampin (RIF) is a first-line antimicrobial agent with potent bactericidal action. Nonetheless, the blood-brain barrier (BBB) limits the therapeutic effects on CNS-TB. Exosomes, however, can facilitate drug movements across the BBB. In addition, exosomes show high biocompatibility and drug-loading capacity. They can also be modified to increase drug delivery efficacy. In this study, we loaded RIF into exosomes and modified the exosomes with a brain-targeting peptide to improve BBB permeability of RIF; we named these exosomes ANG-Exo-RIF. Methods Exosomes were isolated from the culture medium of BMSCs by differential ultracentrifugation and loaded RIF by electroporation and modified ANG by chemical reaction. To characterize ANG-Exo-RIF, Western blot (WB), nanoparticle tracking analysis (NTA) and transmission electron microscopy (TEM) were performed. Bend.3 cells were incubated with DiI labeled ANG-Exo-RIF and then fluorescent microscopy and flow cytometry were used to evaluate the targeting ability of ANG-Exo-RIF in vitro. Fluorescence imaging and frozen section were used to evaluate the targeting ability of ANG-Exo-RIF in vivo. MIC and MBC were determined through microplate alamar blue assay (MABA). Results A novel exosome-based nanoparticle was developed. Compared with untargeted exosomes, the targeted exosomes exhibited high targeting capacity and permeability in vitro and in vivo. The MIC and MBC of ANG-Exo-RIF were 0.25 μg/mL, which were sufficient to meet the clinical needs. Conclusion In summary, excellent targeting ability, high antitubercular activity and biocompatibility endow ANG-Exo-RIF with potential for use in future translation-aimed research and provide hope for an effective CNS-TB treatment.
Collapse
Affiliation(s)
- Han Li
- Department of Tuberculosis, The Second Hospital of Nanjing, Nanjing University of Chinese Medicine, Nanjing, People’s Republic of China
| | - Yinan Ding
- Medical School of Southeast University, Nanjing, People’s Republic of China
| | - Jiayan Huang
- Department of Tuberculosis, The Second Hospital of Nanjing, Nanjing University of Chinese Medicine, Nanjing, People’s Republic of China
| | - Yanyan Zhao
- Department of Tuberculosis, The Second Hospital of Nanjing, Nanjing University of Chinese Medicine, Nanjing, People’s Republic of China
| | - Wei Chen
- Department of Clinical Research Center, The Second Hospital of Nanjing, Nanjing University of Chinese Medicine, Nanjing, People’s Republic of China
| | - Qiusha Tang
- Medical School of Southeast University, Nanjing, People’s Republic of China
| | - Yanli An
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, People’s Republic of China
| | - Rong Chen
- Department of Oncology, Zhongda Hospital, Southeast University, Nanjing, Jiangsu, People’s Republic of China
| | - Chunmei Hu
- Department of Tuberculosis, The Second Hospital of Nanjing, Nanjing University of Chinese Medicine, Nanjing, People’s Republic of China,Correspondence: Chunmei Hu, Department of tuberculosis, the Second Hospital of Nanjing, Nanjing University of Chinese Medicine, Nanjing, 210009, People’s Republic of China, Email
| |
Collapse
|
67
|
Zhang J, Song H, Dong Y, Li G, Li J, Cai Q, Yuan S, Wang Y, Song H. Surface Engineering of HEK293 Cell-Derived Extracellular Vesicles for Improved Pharmacokinetic Profile and Targeted Delivery of IL-12 for the Treatment of Hepatocellular Carcinoma. Int J Nanomedicine 2023; 18:209-223. [PMID: 36660339 PMCID: PMC9844138 DOI: 10.2147/ijn.s388916] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 12/24/2022] [Indexed: 01/14/2023] Open
Abstract
Background Extracellular vesicles (EVs) are considered a promising drug delivery platform. Naïve EVs face numerous issues that limit their applications, such as fast clearance, hepatic accumulations, and a lack of target-specific tropism. We aimed to explore a series of surface engineering approaches to: 1) reduce the non-specific adhesion of EVs, and 2) improve their enrichment in the target tissue. As a proof-of-concept, we investigated the therapeutic potentials of a multi-modal EVs system carrying a tumor-specific nanobody and the immuno-stimulant interleukin-12 (IL12) using in vivo models of hepatocellular carcinoma. Methods The major cell adhesion molecule on the HEK293-derived EVs, integrin β1 (ITGB1), was knocked out (KO) by CRISPR/Cas9-mediated gene editing, followed by deglycosylation to generate ITGB1-Deg EVs for the subsequent pharmacokinetic and biodistribution analyses. ITGB1-Deg EVs were further loaded with glypican-3 (GPC3)-specific nanobody (HN3) and mouse single-chain IL12 (mscIL12) to generate ITGB1-mscIL12+HN3+Deg EVs, for evaluation of tumor tropism and therapeutic potential in a mice model of hepatocellular carcinoma. Results Removal of ITGB1 led to the broad suppression of integrins on the EVs surface, resulting in a decrease in cellular uptake. Deglycosylation of ITGB1- EVs gave rise to inhibition of the EVs uptake by activated RAW264.7 cells. ITGB1 removal did not significantly alter the pharmacokinetic behaviors of HEK293-EVs, whereas the ITGB1-Deg EVs exhibited enhanced systemic exposure with reduced hepatic accumulation. Loading of HN3 conferred the ITGB1-Deg EVs with tumor-specific tropism for both subcutaneous and metastasized tumors in mice. The ITGB1-mscIL12+HN3+Deg EVs activated mouse splenocytes with high potency. Systemic administration of the EVs with the equivalent dose of 1.5µg/kg of exosomal IL12 achieved satisfactory tumor growth inhibition and good tolerability. Conclusion The combinatorial approach of EVs surface engineering conferred HEK293-EVs with reduced non-specific clearance and enhanced tumor targeting efficacy, which constituted an efficient delivery platform for critical cancer therapeutics like IL12.
Collapse
Affiliation(s)
- Jing Zhang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, 102206, People’s Republic of China
| | - Haijing Song
- Emergency Medicine, PLA Strategic Support Force Medical Center, Beijing, 100101, People’s Republic of China
| | - Yanan Dong
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, 102206, People’s Republic of China
| | - Ganghui Li
- China Pharmaceutical University, Nanjing, 211198, People’s Republic of China
| | - Jun Li
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, 102206, People’s Republic of China
| | - Qizhe Cai
- Department of Echocardiography, Beijing Chao Yang Hospital, Capital Medical University, Beijing, 100020, People’s Republic of China
| | - Shoujun Yuan
- Beijing Institute of Radiation Medicine, Beijing, 100850, People’s Republic of China
| | - Yi Wang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, 102206, People’s Republic of China
| | - Haifeng Song
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, 102206, People’s Republic of China
| |
Collapse
|
68
|
Yin X, Jiang LH. Extracellular vesicles: Targeting the heart. Front Cardiovasc Med 2023; 9:1041481. [PMID: 36704471 PMCID: PMC9871562 DOI: 10.3389/fcvm.2022.1041481] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Accepted: 12/16/2022] [Indexed: 01/11/2023] Open
Abstract
Cardiovascular diseases rank the highest incidence and mortality worldwide. As the most common type of cardiovascular disease, myocardial infarction causes high morbidity and mortality. Recent studies have revealed that extracellular vesicles, including exosomes, show great potential as a promising cell-free therapy for the treatment of myocardial infarction. However, low heart-targeting efficiency and short plasma half-life have hampered the clinical translation of extracellular vesicle therapy. Currently, four major types of strategies aiming at enhancing target efficiency have been developed, including modifying EV surface, suppressing non-target absorption, increasing the uptake efficiency of target cells, and utilizing a hydrogel patch. This presented review summarizes the current research aimed at EV heart targeting and discusses the challenges and opportunities in EV therapy, which will be beneficial for the development of effective heart-targeting strategies.
Collapse
Affiliation(s)
- Xin Yin
- Faculty of Life Sciences and Technology, Kunming University of Science and Technology, Kunming, China,Department of Ultrasound, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China,The First People’s Hospital of Yunnan, Kunming, Yunnan, China
| | - Li-Hong Jiang
- Department of Ultrasound, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China,The First People’s Hospital of Yunnan, Kunming, Yunnan, China,*Correspondence: Li-Hong Jiang,
| |
Collapse
|
69
|
Al-Jipouri A, Almurisi SH, Al-Japairai K, Bakar LM, Doolaanea AA. Liposomes or Extracellular Vesicles: A Comprehensive Comparison of Both Lipid Bilayer Vesicles for Pulmonary Drug Delivery. Polymers (Basel) 2023; 15:318. [PMID: 36679199 PMCID: PMC9866119 DOI: 10.3390/polym15020318] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Revised: 12/31/2022] [Accepted: 01/01/2023] [Indexed: 01/11/2023] Open
Abstract
The rapid and non-invasive pulmonary drug delivery (PDD) has attracted great attention compared to the other routes. However, nanoparticle platforms, like liposomes (LPs) and extracellular vesicles (EVs), require extensive reformulation to suit the requirements of PDD. LPs are artificial vesicles composed of lipid bilayers capable of encapsulating hydrophilic and hydrophobic substances, whereas EVs are natural vesicles secreted by cells. Additionally, novel LPs-EVs hybrid vesicles may confer the best of both. The preparation methods of EVs are distinguished from LPs since they rely mainly on extraction and purification, whereas the LPs are synthesized from their basic ingredients. Similarly, drug loading methods into/onto EVs are distinguished whereby they are cell- or non-cell-based, whereas LPs are loaded via passive or active approaches. This review discusses the progress in LPs and EVs as well as hybrid vesicles with a special focus on PDD. It also provides a perspective comparison between LPs and EVs from various aspects (composition, preparation/extraction, drug loading, and large-scale manufacturing) as well as the future prospects for inhaled therapeutics. In addition, it discusses the challenges that may be encountered in scaling up the production and presents our view regarding the clinical translation of the laboratory findings into commercial products.
Collapse
Affiliation(s)
- Ali Al-Jipouri
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, D-45147 Essen, Germany
| | - Samah Hamed Almurisi
- Department of Pharmaceutical Technology, Kulliyyah of Pharmacy, International Islamic University Malaysia, Kuantan 25200, Malaysia
| | - Khater Al-Japairai
- Department of Pharmaceutical Engineering, Faculty of Chemical and Process Engineering Technology, Universiti Malaysia Pahang, Gambang 26300, Malaysia
| | - Latifah Munirah Bakar
- Faculty of Applied Sciences, Universiti Teknologi MARA (UiTM) Selangor, Shah Alam 40450, Malaysia
| | - Abd Almonem Doolaanea
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University College MAIWP International (UCMI), Kuala Lumpur 68100, Malaysia
| |
Collapse
|
70
|
Kluszczynska K, Czyz M. Extracellular Vesicles-Based Cell-Cell Communication in Melanoma: New Perspectives in Diagnostics and Therapy. Int J Mol Sci 2023; 24:ijms24020965. [PMID: 36674479 PMCID: PMC9865538 DOI: 10.3390/ijms24020965] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 12/19/2022] [Accepted: 12/22/2022] [Indexed: 01/06/2023] Open
Abstract
Extracellular vesicles (EVs) are a heterogeneous group of cell-secreted particles that carry cargo of functional biomolecules crucial for cell-to-cell communication with both physiological and pathophysiological consequences. In this review, we focus on evidence demonstrating that the EV-mediated crosstalk between melanoma cells within tumor, between melanoma cells and immune and stromal cells, promotes immune evasion and influences all steps of melanoma development from local progression, pre-metastatic niche formation, to metastatic colonization of distant organs. We also discuss the role of EVs in the development of resistance to immunotherapy and therapy with BRAFV600/MEK inhibitors, and shortly summarize the recent advances on the potential applications of EVs in melanoma diagnostics and therapy.
Collapse
|
71
|
De Sousa KP, Rossi I, Abdullahi M, Ramirez MI, Stratton D, Inal JM. Isolation and characterization of extracellular vesicles and future directions in diagnosis and therapy. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2023; 15:e1835. [PMID: 35898167 PMCID: PMC10078256 DOI: 10.1002/wnan.1835] [Citation(s) in RCA: 99] [Impact Index Per Article: 49.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 06/23/2022] [Accepted: 06/30/2022] [Indexed: 01/31/2023]
Abstract
Extracellular vesicles (EVs) are a unique and heterogeneous class of lipid bilayer nanoparticles secreted by most cells. EVs are regarded as important mediators of intercellular communication in both prokaryotic and eukaryotic cells due to their ability to transfer proteins, lipids and nucleic acids to recipient cells. In addition to their physiological role, EVs are recognized as modulators in pathological processes such as cancer, infectious diseases, and neurodegenerative disorders, providing new potential targets for diagnosis and therapeutic intervention. For a complete understanding of EVs as a universal cellular biological system and its translational applications, optimal techniques for their isolation and characterization are required. Here, we review recent progress in those techniques, from isolation methods to characterization techniques. With interest in therapeutic applications of EVs growing, we address fundamental points of EV-related cell biology, such as cellular uptake mechanisms and their biodistribution in tissues as well as challenges to their application as drug carriers or biomarkers for less invasive diagnosis or as immunogens. This article is categorized under: Diagnostic Tools > Biosensing Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease Therapeutic Approaches and Drug Discovery > Nanomedicine for Infectious Disease.
Collapse
Affiliation(s)
- Karina P. De Sousa
- Bioscience Research Group, School of Life and Medical SciencesUniversity of HertfordshireHertfordshireUK
| | - Izadora Rossi
- School of Human SciencesLondon Metropolitan UniversityLondonUK
- Federal University of ParanáCuritibaBrazil
| | | | - Marcel Ivan Ramirez
- Federal University of ParanáCuritibaBrazil
- Carlos Chagas Institute (ICC)CuritibaBrazil
| | - Dan Stratton
- Open UniversityThe School of Life, Health and Chemical SciencesMilton KeynesUK
| | - Jameel Malhador Inal
- Bioscience Research Group, School of Life and Medical SciencesUniversity of HertfordshireHertfordshireUK
- School of Human SciencesLondon Metropolitan UniversityLondonUK
| |
Collapse
|
72
|
Perez GI, Bachmann MH, Kanada M. In Vivo Analysis of Heterogeneous Extracellular Vesicles Using a Red-Shifted Bioluminescence Resonance Energy Transfer Reporter Protein. Methods Mol Biol 2023; 2668:23-32. [PMID: 37140787 DOI: 10.1007/978-1-0716-3203-1_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Current methods for characterizing the biodistribution of extracellular vesicles (EVs) are not sensitive enough to track EVs in vivo, despite significant advances over the past decade. Commonly used lipophilic fluorescent dyes are convenient, but lack specificity and yield inaccurate spatiotemporal images in the long-term tracking of EVs. In contrast, protein-based fluorescent or bioluminescent EV reporters have more accurately revealed their distribution in cells and mouse models. Here, we describe a red-shifted bioluminescence resonance energy transfer (BRET) EV reporter, PalmReNL, to analyze the trafficking of small EVs (<200 nm; sEVs) and medium/large EVs (>200 nm; m/lEVs) in mice. Its advantages are that (i) background signals in bioluminescence imaging (BLI) are negligible and (ii) the photons PalmReNL emits have spectral wavelengths longer than 600 nm and can more efficiently penetrate tissues than reporters emitting shorter wavelength light.
Collapse
Affiliation(s)
- Gloria I Perez
- Institute for Quantitative Health Science and Engineering (IQ), College of Osteopathic Medicine, Michigan State University, East Lansing, MI, USA
| | - Michael H Bachmann
- Institute for Quantitative Health Science and Engineering (IQ), Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI, USA
| | - Masamitsu Kanada
- Institute for Quantitative Health Science and Engineering (IQ), Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, USA.
| |
Collapse
|
73
|
Zheng W, He R, Liang X, Roudi S, Bost J, Coly P, van Niel G, Andaloussi SEL. Cell-specific targeting of extracellular vesicles through engineering the glycocalyx. J Extracell Vesicles 2022; 11:e12290. [PMID: 36463392 PMCID: PMC9719568 DOI: 10.1002/jev2.12290] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 11/16/2022] [Accepted: 11/23/2022] [Indexed: 12/05/2022] Open
Abstract
Extracellular vesicles (EVs) are promising carriers for the delivery of a variety of chemical and biological drugs. However, their efficacy is limited by the lack of cellular specificity. Available methods to improve the tissue specificity of EVs predominantly rely on surface display of proteins and peptides, largely overlooking the dense glycocalyx that constitutes the outermost layer of EVs. In the present study, we report a reconfigurable glycoengineering strategy that can endogenously display glycans of interest on EV surface. Briefly, EV producer cells are genetically engineered to co-express a glycosylation domain (GD) inserted into the large extracellular loop of CD63 (a well-studied EV scaffold protein) and fucosyltransferase VII (FUT7) or IX (FUT9), so that the engineered EVs display the glycan of interest. Through this strategy, we showcase surface display of two types of glycan ligands, sialyl Lewis X (sLeX) and Lewis X, on EVs and achieve high specificity towards activated endothelial cells and dendritic cells, respectively. Moreover, the endothelial cell-targeting properties of sLeX-EVs were combined with the intrinsic therapeutic effects of mesenchymal stem cells (MSCs), leading to enhanced attenuation of endothelial damage. In summary, this study presents a reconfigurable glycoengineering strategy to produce EVs with strong cellular specificity and highlights the glycocalyx as an exploitable trait for engineering EVs.
Collapse
Affiliation(s)
- Wenyi Zheng
- Biomolecular Medicine, Division of Biomolecular and Cellular Medicine, Department of Laboratory MedicineKarolinska InstitutetHuddingeSweden
- Centre for Allogeneic Stem Cell Transplantation (CAST)Karolinska University HospitalHuddingeSweden
| | - Rui He
- Centre for Allogeneic Stem Cell Transplantation (CAST)Karolinska University HospitalHuddingeSweden
- Experimental Cancer Medicine, Division of Biomolecular and Cellular Medicine, Department of Laboratory MedicineKarolinska InstitutetHuddingeSweden
| | - Xiuming Liang
- Biomolecular Medicine, Division of Biomolecular and Cellular Medicine, Department of Laboratory MedicineKarolinska InstitutetHuddingeSweden
- Centre for Allogeneic Stem Cell Transplantation (CAST)Karolinska University HospitalHuddingeSweden
| | - Samantha Roudi
- Biomolecular Medicine, Division of Biomolecular and Cellular Medicine, Department of Laboratory MedicineKarolinska InstitutetHuddingeSweden
- Centre for Allogeneic Stem Cell Transplantation (CAST)Karolinska University HospitalHuddingeSweden
| | - Jeremy Bost
- Biomolecular Medicine, Division of Biomolecular and Cellular Medicine, Department of Laboratory MedicineKarolinska InstitutetHuddingeSweden
- Centre for Allogeneic Stem Cell Transplantation (CAST)Karolinska University HospitalHuddingeSweden
| | - Pierre‐Michael Coly
- Université de Paris, Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266ParisFrance
- GHU Paris Psychiatrie et NeurosciencesHôpital Sainte AnneParisFrance
| | - Guillaume van Niel
- Université de Paris, Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266ParisFrance
- GHU Paris Psychiatrie et NeurosciencesHôpital Sainte AnneParisFrance
| | - Samir E. L. Andaloussi
- Biomolecular Medicine, Division of Biomolecular and Cellular Medicine, Department of Laboratory MedicineKarolinska InstitutetHuddingeSweden
- Centre for Allogeneic Stem Cell Transplantation (CAST)Karolinska University HospitalHuddingeSweden
- EVOX Therapeutics LimitedOxfordUK
| |
Collapse
|
74
|
Roefs MT, Heusermann W, Brans MAD, Snijders Blok C, Lei Z, Vader P, Sluijter JPG. Evaluation and manipulation of tissue and cellular distribution of cardiac progenitor cell-derived extracellular vesicles. Front Pharmacol 2022; 13:1052091. [PMID: 36506565 PMCID: PMC9729535 DOI: 10.3389/fphar.2022.1052091] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 11/11/2022] [Indexed: 11/25/2022] Open
Abstract
Cardiac progenitor cell-derived extracellular vesicles (CPC-EVs) have been successfully applied via different delivery routes for treating post-myocardial infarction injury in several preclinical models. Hence, understanding the in vivo fate of CPC-EVs after systemic or local, i.e. myocardial, delivery is of utmost importance for the further therapeutic application of CPC-EVs in cardiac repair. Here, we studied the tissue- and cell distribution and retention of CPC-EVs after intramyocardial and intravenous injection in mice by employing different EV labeling and imaging techniques. In contrast to progenitor cells, CPC-EVs demonstrated no immediate flush-out from the heart upon intramyocardial injection and displayed limited distribution to other organs over time, as determined by near-infrared imaging in living animals. By employing CUBIC tissue clearing and light-sheet fluorescent microscopy, we observed CPC-EV migration in the interstitial space of the myocardium shortly after EV injection. Moreover, we demonstrated co-localization with cTnI and CD31-positive cells, suggesting their interaction with various cell types present in the heart. On the contrary, after intravenous injection, most EVs accumulated in the liver. To potentiate such a potential systemic cardiac delivery route, targeting the cardiac endothelium could provide openings for directed CPC-EV therapy. We therefore evaluated whether decorating EVs with targeting peptides (TPs) RGD-4C or CRPPR connected to Lamp2b could enhance EV delivery to endothelial cells. Expression of both TPs enhanced CPC-EV uptake under in vitro continuous flow, but did not affect uptake under static cell culture conditions. Together, these data demonstrate that the route of administration influences CPC-EV biodistribution pattern and suggest that specific TPs could be used to target CPC-EVs to the cardiac endothelium. These insights might lead to a better application of CPC-EV therapeutics in the heart.
Collapse
Affiliation(s)
- Marieke T. Roefs
- Laboratory of Experimental Cardiology, University Medical Center Utrecht, Utrecht, Netherlands
| | | | - Maike A. D. Brans
- Laboratory of Experimental Cardiology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Christian Snijders Blok
- Laboratory of Experimental Cardiology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Zhiyong Lei
- Laboratory of Experimental Cardiology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Pieter Vader
- Laboratory of Experimental Cardiology, University Medical Center Utrecht, Utrecht, Netherlands,CDL Research, University Medical Center Utrecht, Utrecht, Netherlands
| | - Joost P. G. Sluijter
- Laboratory of Experimental Cardiology, University Medical Center Utrecht, Utrecht, Netherlands,Circulatory Health Laboratory, Regenerative Medicine Center, University Medical Center Utrecht, University Utrecht, Utrecht, Netherlands,*Correspondence: Joost P. G. Sluijter,
| |
Collapse
|
75
|
Li W, Wang J, Yin X, Shi H, Sun B, Ji M, Song H, Liu J, Dou Y, Xu C, Jiang X, Li J, Li L, Zhang CY, Zhang Y. Construction of a mouse model that can be used for tissue-specific EV screening and tracing in vivo. Front Cell Dev Biol 2022; 10:1015841. [DOI: 10.3389/fcell.2022.1015841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 11/08/2022] [Indexed: 11/21/2022] Open
Abstract
Extracellular vesicles (EVs) play an important role in the communication between tissues and cells. However, it is difficult to screen and trace EVs secreted by specific tissues in vivo, which affects the functional study of EVs in certain tissues under pathophysiological conditions. In this study, a Cre-dependent CD63flag-EGFP co-expressed with mCherry protein system expressing mice was constructed, which can be used for the secretion, movement, and sorting of EVs from specific tissues in vivo. This mouse model is an ideal research tool for studying the secretion amount, target tissue, and functional molecule screening of EVs in specific tissues under different pathophysiological conditions. Moreover, it provides a new research method to clarify the mechanism of secreted EVs in the pathogenesis of the disease.
Collapse
|
76
|
Liu Q, Huang J, Xia J, Liang Y, Li G. Tracking tools of extracellular vesicles for biomedical research. Front Bioeng Biotechnol 2022; 10:943712. [PMID: 36466335 PMCID: PMC9716315 DOI: 10.3389/fbioe.2022.943712] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Accepted: 11/03/2022] [Indexed: 08/02/2023] Open
Abstract
Imaging of extracellular vesicles (EVs) will facilitate a better understanding of their biological functions and their potential as therapeutics and drug delivery vehicles. In order to clarify EV-mediated cellular communication in vitro and to track the bio-distribution of EV in vivo, various strategies have been developed to label and image EVs. In this review, we summarized recent advances in the tracking of EVs, demonstrating the methods for labeling and imaging of EVs, in which the labeling methods include direct and indirect labeling and the imaging modalities include fluorescent imaging, bioluminescent imaging, nuclear imaging, and nanoparticle-assisted imaging. These techniques help us better understand the mechanism of uptake, the bio-distribution, and the function of EVs. More importantly, we can evaluate the pharmacokinetic properties of EVs, which will help promote their further clinical application.
Collapse
Affiliation(s)
- Qisong Liu
- Shenzhen Key Laboratory of Musculoskeletal Tissue Reconstruction and Function Restoration, Department of Orthopaedic Surgery, Shenzhen People’s Hospital (The Second Clinical Medical College of Jinan University), Shenzhen, China
| | - Jianghong Huang
- Department of Orthopedics, Shenzhen Second People’s Hospital (First Affiliated Hospital of Shenzhen University, Health Science Center), Shenzhen, China
- Tsinghua University Shenzhen International Graduate School, Shenzhen, China
| | - Jiang Xia
- Department of Chemistry, The Chinese University of Hong Kong, Hong Kong, China
| | - Yujie Liang
- Department of Child and Adolescent Psychiatry, Shenzhen Kangning Hospital, Shenzhen Mental Health Center, Shenzhen, China
- Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, China
| | - Guangheng Li
- Shenzhen Key Laboratory of Musculoskeletal Tissue Reconstruction and Function Restoration, Department of Orthopaedic Surgery, Shenzhen People’s Hospital (The Second Clinical Medical College of Jinan University), Shenzhen, China
| |
Collapse
|
77
|
Pauwels MJ, Xie J, Ceroi A, Balusu S, Castelein J, Van Wonterghem E, Van Imschoot G, Ward A, Menheniott TR, Gustafsson O, Combes F, El Andaloussi S, Sanders NN, Mäger I, Van Hoecke L, Vandenbroucke RE. Choroid plexus-derived extracellular vesicles exhibit brain targeting characteristics. Biomaterials 2022; 290:121830. [PMID: 36302306 DOI: 10.1016/j.biomaterials.2022.121830] [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: 05/11/2022] [Revised: 09/16/2022] [Accepted: 09/25/2022] [Indexed: 11/17/2022]
Abstract
The brain is protected against invading organisms and other unwanted substances by tightly regulated barriers. However, these central nervous system (CNS) barriers impede the delivery of drugs into the brain via the blood circulation and are therefore considered major hurdles in the treatment of neurological disorders. Consequently, there is a high need for efficient delivery systems that are able to cross these strict barriers. While most research focuses on the blood-brain barrier (BBB), the design of drug delivery platforms that are able to cross the blood-cerebrospinal fluid (CSF) barrier, formed by a single layer of choroid plexus epithelial cells, remains a largely unexplored domain. The discovery that extracellular vesicles (EVs) make up a natural mechanism for information transfer between cells and across cell layers, has stimulated interest in their potential use as drug delivery platform. Here, we report that choroid plexus epithelial cell-derived EVs exhibit the capacity to home to the brain after peripheral administration. Moreover, these vesicles are able to functionally deliver cargo into the brain. Our findings underline the therapeutic potential of choroid plexus-derived EVs as a brain drug delivery vehicle via targeting of the blood-CSF interface.
Collapse
Affiliation(s)
- Marie J Pauwels
- VIB Center for Inflammation Research, VIB, Technologiepark-Zwijnaarde 71, 9052, Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Technologiepark-Zwijnaarde 71, 9052, Ghent, Belgium
| | - Junhua Xie
- VIB Center for Inflammation Research, VIB, Technologiepark-Zwijnaarde 71, 9052, Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Technologiepark-Zwijnaarde 71, 9052, Ghent, Belgium
| | - Adam Ceroi
- VIB Center for Inflammation Research, VIB, Technologiepark-Zwijnaarde 71, 9052, Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Technologiepark-Zwijnaarde 71, 9052, Ghent, Belgium
| | - Sriram Balusu
- VIB Center for the Biology of Disease, VIB, Herestraat 49, 3000, Leuven, Belgium
| | - Jonas Castelein
- VIB Center for Inflammation Research, VIB, Technologiepark-Zwijnaarde 71, 9052, Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Technologiepark-Zwijnaarde 71, 9052, Ghent, Belgium
| | - Elien Van Wonterghem
- VIB Center for Inflammation Research, VIB, Technologiepark-Zwijnaarde 71, 9052, Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Technologiepark-Zwijnaarde 71, 9052, Ghent, Belgium
| | - Griet Van Imschoot
- VIB Center for Inflammation Research, VIB, Technologiepark-Zwijnaarde 71, 9052, Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Technologiepark-Zwijnaarde 71, 9052, Ghent, Belgium
| | - Andrew Ward
- Department of Biology and Biochemistry, University of Bath, Claverton Down, Bath, BA2 7AY, UK
| | - Trevelyan R Menheniott
- Murdoch Children's Research Institute, Flemington Rd. Parkville, Melbourne, Victoria, Australia; Department of Paediatrics, University of Melbourne, Flemington Rd. Parkville, Melbourne, Victoria, Australia
| | - Oskar Gustafsson
- Department of Laboratory Medicine, Karolinska Institutet, 141 86 Stockholm, Sweden
| | - Francis Combes
- Department of Biotechnology and Nanomedicine, SINTEF AS, Sem Sælands V. 2A, N-7034 Trondheim, Norway
| | - Samir El Andaloussi
- Department of Laboratory Medicine, Karolinska Institutet, 141 86 Stockholm, Sweden
| | - Niek N Sanders
- Laboratory of Gene Therapy, Department of Nutrition, Genetics and Ethology, Faculty of Veterinary Medicine, Ghent University, Heidestraat 19, 9820 Merelbeke, Belgium; Cancer Research Institute Ghent (CRIG), 9000, Ghent, Belgium
| | - Imre Mäger
- Institute of Technology, University of Tartu, 50 411, Tartu, Estonia; Department of Paediatrics, University of Oxford, Oxford, OX3 9DU, UK
| | - Lien Van Hoecke
- VIB Center for Inflammation Research, VIB, Technologiepark-Zwijnaarde 71, 9052, Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Technologiepark-Zwijnaarde 71, 9052, Ghent, Belgium
| | - Roosmarijn E Vandenbroucke
- VIB Center for Inflammation Research, VIB, Technologiepark-Zwijnaarde 71, 9052, Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Technologiepark-Zwijnaarde 71, 9052, Ghent, Belgium.
| |
Collapse
|
78
|
Komuro H, Aminova S, Lauro K, Harada M. Advances of engineered extracellular vesicles-based therapeutics strategy. SCIENCE AND TECHNOLOGY OF ADVANCED MATERIALS 2022; 23:655-681. [PMID: 36277506 PMCID: PMC9586594 DOI: 10.1080/14686996.2022.2133342] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 09/26/2022] [Accepted: 09/28/2022] [Indexed: 05/09/2023]
Abstract
Extracellular vesicles (EVs) are a heterogeneous population of lipid bilayer membrane-bound vesicles which encapsulate bioactive molecules, such as nucleic acids, proteins, and lipids. They mediate intercellular communication through transporting internally packaged molecules, making them attractive therapeutics carriers. Over the last decades, a significant amount of research has implied the potential of EVs servings as drug delivery vehicles for nuclear acids, proteins, and small molecular drugs. However, several challenges remain unresolved before the clinical application of EV-based therapeutics, including lack of specificity, stability, biodistribution, storage, large-scale manufacturing, and the comprehensive analysis of EV composition. Technical development is essential to overcome these issues and enhance the pre-clinical therapeutic effects. In this review, we summarize the current advancements in EV engineering which demonstrate their therapeutic potential.
Collapse
Affiliation(s)
- Hiroaki Komuro
- Institute for Quantitative Health Science and Engineering (IQ), Michigan State University, East Lansing, MI, USA
- Department of Biomedical Engineering, Michigan State University, East Lansing, MI, USA
| | - Shakhlo Aminova
- Institute for Quantitative Health Science and Engineering (IQ), Michigan State University, East Lansing, MI, USA
- Department of Biomedical Engineering, Michigan State University, East Lansing, MI, USA
| | - Katherine Lauro
- Institute for Quantitative Health Science and Engineering (IQ), Michigan State University, East Lansing, MI, USA
- Department of Biomedical Engineering, Michigan State University, East Lansing, MI, USA
| | - Masako Harada
- Institute for Quantitative Health Science and Engineering (IQ), Michigan State University, East Lansing, MI, USA
- Department of Biomedical Engineering, Michigan State University, East Lansing, MI, USA
| |
Collapse
|
79
|
Characteristics of Exosomes and the Vascular Landscape Regulate Exosome Sequestration by Peripheral Tissues and Brain. Int J Mol Sci 2022; 23:ijms232012513. [PMID: 36293369 PMCID: PMC9603979 DOI: 10.3390/ijms232012513] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 10/05/2022] [Accepted: 10/12/2022] [Indexed: 02/04/2023] Open
Abstract
Exosomes mediate intercellular communication, shuttling messages between cells and tissues. We explored whether exosome tissue sequestration is determined by the exosomes or the tissues using ten radiolabeled exosomes from human or murine, cancerous or noncancerous cell lines. We measured sequestration of these exosomes by the liver, kidney, spleen, and lung after intravenous injection into male CD-1 mice. Except for kidney sequestration of three exosomes, all exosomes were incorporated by all tissues, but sequestration levels varied greatly among exosomes and tissues. Species of origin (mouse vs. human) or source (cancerous vs. noncancerous cells) did not influence tissue sequestration. Sequestration of J774A.1 exosomes by liver involved the mannose-6 phosphate (M6P) receptor. Wheatgerm agglutinin (WGA) or lipopolysaccharide (LPS) treatments enhanced sequestration of exosomes by brain and lung but inhibited sequestration by liver and spleen. Response to LPS was not predictive of response to WGA. Path and heat map analyses included our published results for brain and found distinct clusters among the exosomes and the tissues. In conclusion, we found no evidence for a universal binding site controlling exosome-tissue interactions. Instead, sequestration of exosomes by tissues is differentially regulated by both exosomes and tissues and may be stimulated or inhibited by WGA and inflammation.
Collapse
|
80
|
Macromolecular assembly of bioluminescent protein nanoparticles for enhanced imaging. Mater Today Bio 2022; 17:100455. [PMID: 36304975 PMCID: PMC9593766 DOI: 10.1016/j.mtbio.2022.100455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 10/05/2022] [Accepted: 10/07/2022] [Indexed: 11/11/2022] Open
Abstract
Bioluminescence imaging has advantages over fluorescence imaging, such as minimal photobleaching and autofluorescence, and greater signal-to-noise ratios in many complex environments. Although significant achievements have been made in luciferase engineering for generating bright and stable reporters, the full capability of luciferases for nanoparticle tracking has not been comprehensively examined. In biocatalysis, enhanced enzyme performance after immobilization on nanoparticles has been reported. Thus, we hypothesized that by assembling luciferases onto a nanoparticle, the resulting complex could lead to substantially improved imaging properties. Using a modular bioconjugation strategy, we attached NanoLuc (NLuc) or Akaluc bioluminescent proteins to a protein nanoparticle platform (E2), yielding nanoparticles NLuc-E2 and Akaluc-E2, both with diameters of ∼45 nm. Although no significant differences were observed between different conditions involving Akaluc and Akaluc-E2, free NLuc at pH 5.0 showed significantly lower emission values than free NLuc at pH 7.4. Interestingly, NLuc immobilization on E2 nanoparticles (NLuc-E2) emitted increased luminescence at pH 7.4, and at pH 5.0 showed over two orders of magnitude (>200-fold) higher luminescence (than free NLuc), expanding the potential for imaging detection using the nanoparticle even upon endocytic uptake. After uptake by macrophages, the resulting luminescence with NLuc-E2 nanoparticles was up to 7-fold higher than with free NLuc at 48 h. Cells incubated with NLuc-E2 could also be imaged using live bioluminescence microscopy. Finally, biodistribution of nanoparticles into lymph nodes was detected through imaging using NLuc-E2, but not with conventionally-labeled fluorescent E2. Our data demonstrate that NLuc-bound nanoparticles have advantageous properties that can be utilized in applications ranging from single-cell imaging to in vivo biodistribution.
Collapse
|
81
|
Hirose H, Hirai Y, Sasaki M, Sawa H, Futaki S. Quantitative Analysis of Extracellular Vesicle Uptake and Fusion with Recipient Cells. Bioconjug Chem 2022; 33:1852-1859. [PMID: 36194183 DOI: 10.1021/acs.bioconjchem.2c00307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
In precision medicine, extracellular vesicles (EVs) are promising intracellular drug delivery vehicles. The development of a quantitative analysis approach will provide valuable information from the perspective of cell biology and system design for drug delivery. Previous studies have reported quantitative methods to analyze the relative uptake or fusion of EVs to recipient cells. However, relatively few methods have enabled the simultaneous evaluation of the "number" of EVs taken up by recipient cells and those that fuse with cellular membranes. In this study, we report a simple quantitative method based on the NanoBiT system to quantify the uptake and fusion of small and large EVs (sEVs and lEVs, respectively). We assessed the abundance of these two subtypes of EVs and determined that lEVs may be more effective vehicles for transporting cargo to recipient cells. The results also indicated that both sEVs and lEVs have very low fusogenic activity, which can be improved in the presence of a fusogenic protein.
Collapse
Affiliation(s)
- Hisaaki Hirose
- Institute for Chemical Research, Kyoto University, Uji, Kyoto611-0011, Japan
| | - Yusuke Hirai
- Institute for Chemical Research, Kyoto University, Uji, Kyoto611-0011, Japan
| | - Michihito Sasaki
- Division of Molecular Pathobiology, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Hokkaido001-0020, Japan
| | - Hirofumi Sawa
- Division of Molecular Pathobiology, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Hokkaido001-0020, Japan
| | - Shiroh Futaki
- Institute for Chemical Research, Kyoto University, Uji, Kyoto611-0011, Japan
| |
Collapse
|
82
|
Driedonks T, Jiang L, Carlson B, Han Z, Liu G, Queen SE, Shirk EN, Gololobova O, Liao Z, Nyberg LH, Lima G, Paniushkina L, Garcia‐Contreras M, Schonvisky K, Castell N, Stover M, Guerrero‐Martin S, Richardson R, Smith B, Mahairaki V, Lai CP, Izzi JM, Hutchinson EK, Pate KAM, Witwer KW. Pharmacokinetics and biodistribution of extracellular vesicles administered intravenously and intranasally to Macaca nemestrina. JOURNAL OF EXTRACELLULAR BIOLOGY 2022; 1:e59. [PMID: 36591537 PMCID: PMC9799283 DOI: 10.1002/jex2.59] [Citation(s) in RCA: 70] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Revised: 07/23/2022] [Accepted: 08/13/2022] [Indexed: 01/05/2023]
Abstract
Extracellular vesicles (EVs) have potential in disease treatment since they can be loaded with therapeutic molecules and engineered for retention by specific tissues. However, questions remain on optimal dosing, administration, and pharmacokinetics. Previous studies have addressed biodistribution and pharmacokinetics in rodents, but little evidence is available for larger animals. Here, we investigated the pharmacokinetics and biodistribution of Expi293F-derived EVs labelled with a highly sensitive nanoluciferase reporter (palmGRET) in a non-human primate model (Macaca nemestrina), comparing intravenous (IV) and intranasal (IN) administration over a 125-fold dose range. We report that EVs administered IV had longer circulation times in plasma than previously reported in mice and were detectable in cerebrospinal fluid (CSF) after 30-60 minutes. EV association with PBMCs, especially B-cells, was observed as early as one minute post-administration. EVs were detected in liver and spleen within one hour of IV administration. However, IN delivery was minimal, suggesting that pretreatment approaches may be needed in large animals. Furthermore, EV circulation times strongly decreased after repeated IV administration, possibly due to immune responses and with clear implications for xenogeneic EV-based therapeutics. We hope that our findings from this baseline study in macaques will help to inform future research and therapeutic development of EVs.
Collapse
Affiliation(s)
- Tom Driedonks
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Linglei Jiang
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Bess Carlson
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Zheng Han
- Russell H. Morgan Department of RadiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
- F.M. Kirby Research CenterKennedy Krieger InstituteBaltimoreMarylandUSA
| | - Guanshu Liu
- Russell H. Morgan Department of RadiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
- F.M. Kirby Research CenterKennedy Krieger InstituteBaltimoreMarylandUSA
| | - Suzanne E. Queen
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Erin N. Shirk
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Olesia Gololobova
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Zhaohao Liao
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Lyle H. Nyberg
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Gabriela Lima
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Liliia Paniushkina
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Marta Garcia‐Contreras
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Kayla Schonvisky
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Natalie Castell
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Mitchel Stover
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Selena Guerrero‐Martin
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
- Division of Comparative Medicine and Department of Biological EngineeringMassachusetts Institute of TechnologyCambridgeMassachusettsUSA
| | - Riley Richardson
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Barbara Smith
- Department of Cell BiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Vasiliki Mahairaki
- The Richman Family Precision Medicine Center of Excellence in Alzheimer's DiseaseJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Charles P. Lai
- Institute of Atomic and Molecular SciencesAcademia SinicaTaipeiTaiwan
| | - Jessica M. Izzi
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Eric K. Hutchinson
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Kelly A. M. Pate
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
- Division of Comparative Medicine and Department of Biological EngineeringMassachusetts Institute of TechnologyCambridgeMassachusettsUSA
| | - Kenneth W. Witwer
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
- Department of NeurologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
- The Richman Family Precision Medicine Center of Excellence in Alzheimer's DiseaseJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| |
Collapse
|
83
|
Aimaletdinov AM, Gomzikova MO. Tracking of Extracellular Vesicles' Biodistribution: New Methods and Approaches. Int J Mol Sci 2022; 23:11312. [PMID: 36232613 PMCID: PMC9569979 DOI: 10.3390/ijms231911312] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 09/15/2022] [Accepted: 09/21/2022] [Indexed: 12/12/2022] Open
Abstract
Extracellular vesicles (EVs) are nanosized lipid bilayer vesicles that are released by almost all cell types. They range in diameter from 30 nm to several micrometres and have the ability to carry biologically active molecules such as proteins, lipids, RNA, and DNA. EVs are natural vectors and play an important role in many physiological and pathological processes. The amount and composition of EVs in human biological fluids serve as biomarkers and are used for diagnosing diseases and monitoring the effectiveness of treatment. EVs are promising for use as therapeutic agents and as natural vectors for drug delivery. However, the successful use of EVs in clinical practice requires an understanding of their biodistribution in an organism. Numerous studies conducted so far on the biodistribution of EVs show that, after intravenous administration, EVs are mostly localized in organs rich in blood vessels and organs associated with the reticuloendothelial system, such as the liver, lungs, spleen, and kidneys. In order to improve resolution, new dyes and labels are being developed and detection methods are being optimized. In this work, we review all available modern methods and approaches used to assess the biodistribution of EVs, as well as discuss their advantages and limitations.
Collapse
Affiliation(s)
| | - Marina O. Gomzikova
- Laboratory of Intercellular Communication, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan 420008, Russia
| |
Collapse
|
84
|
Wan T, Zhong J, Pan Q, Zhou T, Ping Y, Liu X. Exosome-mediated delivery of Cas9 ribonucleoprotein complexes for tissue-specific gene therapy of liver diseases. SCIENCE ADVANCES 2022; 8:eabp9435. [PMID: 36103526 PMCID: PMC9473578 DOI: 10.1126/sciadv.abp9435] [Citation(s) in RCA: 138] [Impact Index Per Article: 46.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/13/2023]
Abstract
CRISPR-Cas9 gene editing has emerged as a powerful therapeutic technology, but the lack of safe and efficient in vivo delivery systems, especially for tissue-specific vectors, limits its broad clinical applications. Delivery of Cas9 ribonucleoprotein (RNP) owns competitive advantages over other options; however, the large size of RNPs exceeds the loading capacity of currently available delivery vectors. Here, we report a previously unidentified genome editing delivery system, named exosomeRNP, in which Cas9 RNPs were loaded into purified exosomes isolated from hepatic stellate cells through electroporation. ExosomeRNP facilitated effective cytosolic delivery of RNP in vitro while specifically accumulated in the liver tissue in vivo. ExosomeRNP showed vigorous therapeutic potential in acute liver injury, chronic liver fibrosis, and hepatocellular carcinoma mouse models via targeting p53 up-regulated modulator of apoptosis (PUMA), cyclin E1 (CcnE1), and K (lysine) acetyltransferase 5 (KAT5), respectively. The developed exosomeRNP provides a feasible platform for precise and tissue-specific gene therapies of liver diseases.
Collapse
Affiliation(s)
- Tao Wan
- Liangzhu Laboratory, Zhejiang University Medical Center, Zhejiang University, Hangzhou 311121, China
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Jiafeng Zhong
- Department of Pharmacology and Department of Gastroenterology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310027, China
| | - Qi Pan
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Tianhua Zhou
- Liangzhu Laboratory, Zhejiang University Medical Center, Zhejiang University, Hangzhou 311121, China
- Cancer Center, Zhejiang University, Hangzhou 310058, China
- Department of Cell Biology, School of Medicine, Zhejiang University, Hangzhou 310058, China
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
- Corresponding author. (X.L.); (Y.P.); (T.Z.)
| | - Yuan Ping
- Liangzhu Laboratory, Zhejiang University Medical Center, Zhejiang University, Hangzhou 311121, China
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Corresponding author. (X.L.); (Y.P.); (T.Z.)
| | - Xiangrui Liu
- Liangzhu Laboratory, Zhejiang University Medical Center, Zhejiang University, Hangzhou 311121, China
- Department of Pharmacology and Department of Gastroenterology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
- Cancer Center, Zhejiang University, Hangzhou 310058, China
- Corresponding author. (X.L.); (Y.P.); (T.Z.)
| |
Collapse
|
85
|
Dhar R, Mukherjee S, Mukerjee N, Mukherjee D, Devi A, Ashraf GM, Alserihi RF, Tayeb HH, Hashem AM, Alexiou A, Thorate N. Interrelation between extracellular vesicles miRNAs with chronic lung diseases. J Cell Physiol 2022; 237:4021-4036. [PMID: 36063496 DOI: 10.1002/jcp.30867] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 08/18/2022] [Accepted: 08/22/2022] [Indexed: 11/07/2022]
Abstract
Extracellular vehicles (EVs) are nanoscale lipid bilayer vesicles that carry biologically active biomolecule cargos like proteins, lipids, and nucleic acids (DNA, RNA) outside of the cell. Blood (serum/plasma), urine, and bronchoalveolar lavage fluid are all examples of biofluids from which they may be collected. EVs play a vital role in intracellular communication. The molecular signature of EVs largely depends on the parental cell's status. EVs are classified into two groups, (1) exosomes (originated by endogenous route) and (2) microvesicles (originated from the plasma membrane, also known as ectosomes). The quantity and types of EV cargo vary during normal conditions compared to pathological conditions (chronic inflammatory lung diseases or lung cancer). Consequently, EVs contain novel biomarkers that differ based on the cell type of origin and during lung diseases. Small RNAs (e.g., microRNAs) are transported by EVs, which is one of the most rapidly evolving research areas in the field of EVs biology. EV-mediated cargos transport small RNAs that can result in reprograming the target/recipient cells. Multiple chronic inflammatory lung illnesses, such as chronic obstructive pulmonary disease, asthma, pulmonary hypertension, pulmonary fibrosis, cystic fibrosis, acute lung injury, and lung cancer, have been demonstrated to be regulated by EV. In this review, we will consolidate the current knowledge and literature on the novel role of EVs and their small RNAs concerning chronic lung diseases (CLDs). Additionally, we will also provide better insight into the clinical and translational impact of mesenchymal stem cells-derived EVs as novel therapeutic agents in treating CLDs.
Collapse
Affiliation(s)
- Rajib Dhar
- Cancer and Stem Cell Biology Laboratory, Department of Genetic Engineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India
| | - Sayantanee Mukherjee
- Centre for Nanosciences and Molecular Medicine, Amrita Vishwa Vidyapeetham University, Kochi, Kerala, India
| | - Nobendu Mukerjee
- Department of Microbiology, Ramakrishna Mission Vivekananda Centenary College, Kolkata, West Bengal, India.,Department of Health Sciences, Novel Global Community Educational Foundation, Hebersham, Australia
| | | | - Arikketh Devi
- Cancer and Stem Cell Biology Laboratory, Department of Genetic Engineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India
| | - Ghulam Md Ashraf
- Pre-Clinical Research Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia.,Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Raed F Alserihi
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia.,Hematology Research Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia.,Nanomedicine Unit, Center of Innovation in Personalized Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Hossam H Tayeb
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia.,Nanomedicine Unit, Center of Innovation in Personalized Medicine, King Abdulaziz University, Jeddah, Saudi Arabia.,King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Anwar M Hashem
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia.,Vaccines and Immunotherapy Unit, King Fahad Medical Research Center, Jeddah, Saudi Arabia
| | - Athanasios Alexiou
- Department of Science and Engineering, Novel Global Community Educational Foundation, Hebersham, Australia
| | - Nanasaheb Thorate
- Division of Medical Sciences, Nuffield Department of Women's & Reproductive Health, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
86
|
Lu Y, Wang L, Zhang M, Chen Z. Mesenchymal Stem Cell-Derived Small Extracellular Vesicles: A Novel Approach for Kidney Disease Treatment. Int J Nanomedicine 2022; 17:3603-3618. [PMID: 35990308 PMCID: PMC9386173 DOI: 10.2147/ijn.s372254] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 08/04/2022] [Indexed: 12/24/2022] Open
Abstract
Globally, kidney disease has become a serious health challenge, with approximately 10% of adults suffering with the disease, and increasing incidence and mortality rates every year. Small extracellular vesicles (sEVs) are 30 nm-100 nm sized nanovesicles released by cells into the extracellular matrix (ECM), which serve as mediators of intercellular communication. Depending on the cell origin, sEVs have different roles which depend on internal cargoes including, nucleic acids, proteins, and lipids. Mesenchymal stem cell (MSCs) exert anti-inflammatory, anti-aging, and wound healing functions mainly via sEVs in a stable and safe manner. MSC-derived sEVs (MSC-sEVs) exert roles in several kidney diseases by transporting renoprotective cargoes to reduce oxidative stress, inhibit renal cell apoptosis, suppress inflammation, and mediate anti-fibrosis mechanisms. Additionally, because MSC-sEVs efficiently target damaged kidneys, they have the potential to become the next generation cell-free therapies for kidney disease. Herein, we review recent research data on how MSC-sEVs could be used to treat kidney disease.
Collapse
Affiliation(s)
- Yukang Lu
- First Clinical Medical College, Gannan Medical University, Ganzhou, People's Republic of China.,Department of Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, People's Republic of China
| | - Lanfeng Wang
- Department of Nephrology, First Affiliated Hospital of Gannan Medical University, Ganzhou, People's Republic of China
| | - Mengting Zhang
- First Clinical Medical College, Gannan Medical University, Ganzhou, People's Republic of China.,Department of Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, People's Republic of China
| | - Zhiping Chen
- Department of Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, People's Republic of China
| |
Collapse
|
87
|
Cancer associated fibroblasts secreted exosomal miR-1290 contributes to prostate cancer cell growth and metastasis via targeting GSK3β. Cell Death Dis 2022; 8:371. [PMID: 35999213 PMCID: PMC9399109 DOI: 10.1038/s41420-022-01163-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 08/08/2022] [Accepted: 08/10/2022] [Indexed: 01/22/2023]
Abstract
Cancer-associated fibroblasts (CAFs) play crucial roles in mediating tumor growth and metastasis via transferring exosomes to neighboring cells, whereas the mechanisms by which CAFs regulate the tumorgenesis of prostate cancer (PC) remain largely unknown. In this study, CAFs and normal fibroblasts (NFs) were isolated from PC tissues and adjacent normal tissues, respectively. Exosomes (NFs-Exo and CAFs-Exo) were then isolated from the supernatant of NFs and CAFs. Next, the differentially expressed miRNAs (DEMs) between NFs-Exo and CAFs-Exo were identified using RNA-sequencing. Cell viability, migration and invasion were detected with CCK-8 and Transwell assays. Protein expression was measured with western blot. We found that CAFs-Exo remarkably enhanced PC cell migration, invasion, stemness, epithelial-mesenchymal transition (EMT) and metastasis. Significantly, miR-1290 level was upregulated in CAFs-Exo compared to NFs-Exo. In addition, CAFs could transfer exosomes to PC cells, resulting in a marked increase of miR-1290 level in cells. Moreover, exosomal miR-1290 could inhibit GSK3β/β-catenin signaling by binding with the downstream target GSK3β mRNA. Meanwhile, miR-1290 antagomir notably reversed the effects of CAFs-Exo on PC cells through activating GSK3β/β-catenin signaling. Collectively, exosomal miR-1290 from CAFs could promote PC cell growth and metastasis via inhibiting GSK3β/β-catenin signaling, suggesting that miR-1290 may serve as potential therapeutic target for the treatment of PC.
Collapse
|
88
|
Liang X, Niu Z, Galli V, Howe N, Zhao Y, Wiklander OPB, Zheng W, Wiklander RJ, Corso G, Davies C, Hean J, Kyriakopoulou E, Mamand DR, Amin R, Nordin JZ, Gupta D, Andaloussi SEL. Extracellular vesicles engineered to bind albumin demonstrate extended circulation time and lymph node accumulation in mouse models. J Extracell Vesicles 2022; 11:e12248. [PMID: 35879268 PMCID: PMC9314316 DOI: 10.1002/jev2.12248] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 06/29/2022] [Accepted: 07/11/2022] [Indexed: 11/21/2022] Open
Abstract
Extracellular vesicles (EVs) have shown promise as potential therapeutics for the treatment of various diseases. However, their rapid clearance after administration could be a limitation in certain therapeutic settings. To solve this, an engineering strategy is employed to decorate albumin onto the surface of the EVs through surface display of albumin binding domains (ABDs). ABDs were either included in the extracellular loops of select EV-enriched tetraspanins (CD63, CD9 and CD81) or directly fused to the extracellular terminal of single transmembrane EV-sorting domains, such as Lamp2B. These engineered EVs exert robust binding capacity to human serum albumins (HSA) in vitro and mouse serum albumins (MSA) after injection in mice. By binding to MSA, circulating time of EVs dramatically increases after different routes of injection in different strains of mice. Moreover, these engineered EVs show considerable lymph node (LN) and solid tumour accumulation, which can be utilized when using EVs for immunomodulation, cancer- and/or immunotherapy. The increased circulation time of EVs may also be important when combined with tissue-specific targeting ligands and could provide significant benefit for their therapeutic use in a variety of disease indications.
Collapse
Affiliation(s)
- Xiuming Liang
- Biomolecular MedicineClinical Research CenterDepartment of Laboratory Medicine Karolinska InstitutetStockholmSweden
- Cancer Research LaboratoryShandong University‐Karolinska Institutet collaborative LaboratorySchool of Basic Medical ScienceShandong UniversityJinanShandongPR China
| | - Zheyu Niu
- Biomolecular MedicineClinical Research CenterDepartment of Laboratory Medicine Karolinska InstitutetStockholmSweden
- Department of Hepatobiliary SurgeryShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanChina
| | | | | | - Ying Zhao
- Experimental Cancer MedicineClinical Research CenterDepartment of Laboratory MedicineKarolinska InstitutetStockholmSweden
| | - Oscar P. B. Wiklander
- Biomolecular MedicineClinical Research CenterDepartment of Laboratory Medicine Karolinska InstitutetStockholmSweden
| | - Wenyi Zheng
- Biomolecular MedicineClinical Research CenterDepartment of Laboratory Medicine Karolinska InstitutetStockholmSweden
| | - Rim Jawad Wiklander
- Biomolecular MedicineClinical Research CenterDepartment of Laboratory Medicine Karolinska InstitutetStockholmSweden
| | - Giulia Corso
- Biomolecular MedicineClinical Research CenterDepartment of Laboratory Medicine Karolinska InstitutetStockholmSweden
| | | | | | | | - Doste R. Mamand
- Biomolecular MedicineClinical Research CenterDepartment of Laboratory Medicine Karolinska InstitutetStockholmSweden
| | - Risul Amin
- Biomolecular MedicineClinical Research CenterDepartment of Laboratory Medicine Karolinska InstitutetStockholmSweden
| | - Joel Z. Nordin
- Biomolecular MedicineClinical Research CenterDepartment of Laboratory Medicine Karolinska InstitutetStockholmSweden
| | - Dhanu Gupta
- Biomolecular MedicineClinical Research CenterDepartment of Laboratory Medicine Karolinska InstitutetStockholmSweden
| | - Samir EL Andaloussi
- Biomolecular MedicineClinical Research CenterDepartment of Laboratory Medicine Karolinska InstitutetStockholmSweden
- Evox Therapeutics LimitedOxfordUK
| |
Collapse
|
89
|
Affinity-based isolation of extracellular vesicles and the effects on downstream molecular analysis. Anal Bioanal Chem 2022; 414:7051-7067. [PMID: 35732746 DOI: 10.1007/s00216-022-04178-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Revised: 05/25/2022] [Accepted: 06/13/2022] [Indexed: 11/01/2022]
Abstract
Extracellular vesicles (EVs) are transport vesicles with diameters ranging from 30 to 1000 nm, secreted by cells in both physiological and pathological conditions. By using the EV shuttling system, biomolecular cargo such as proteins and genetic materials travels between cells resulting in intercellular communication and epigenetic regulation. Because the presence of EVs and cargo molecules in body fluids can predict the state of the parental cells, EV isolation techniques from complex biofluids have been developed. Further exploration of EVs through downstream molecular analysis depends heavily on those isolation technologies. Methodologies based either on physical separation or on affinity binding have been used to isolate EVs. Affinity-based methods for EV isolation are known to produce highly specific and efficient isolation results. However, so far, there is a lack of literature summarizing these methods and their effects on downstream EV molecular analysis. In the present work, we reviewed recent efforts on developing affinity-based methods for the isolation of EVs, with an emphasis on comparing their effects on downstream analysis of EV molecular cargo. Antibody-based isolation techniques produce highly pure EVs, but the harsh eluents damage the EV structure, and some antibodies stay bound to the EVs after elution. Aptamer-based methods use relatively mild elution conditions and release EVs in their native form, but their isolation efficiencies need to be improved. The membrane affinity-based method and other affinity-based methods based on the properties of the EV lipid bilayer also isolate intact EVs, but they can also result in contaminants. From the perspective of affinity-based methods, we investigated the influence of the isolation methods of choice on downstream EV molecular analysis.
Collapse
|
90
|
Han C, Qin G. Reporter Systems for Assessments of Extracellular Vesicle Transfer. Front Cardiovasc Med 2022; 9:922420. [PMID: 35722089 PMCID: PMC9198260 DOI: 10.3389/fcvm.2022.922420] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Accepted: 05/06/2022] [Indexed: 12/12/2022] Open
Abstract
Extracellular vesicles (EVs) are lipid bilayer particles naturally released from most if not all cell types to mediate inter-cellular exchange of bioactive molecules. Mounting evidence suggest their important role in diverse pathophysiological processes in the development, growth, homeostasis, and disease. Thus, sensitive and reliable assessments of functional EV cargo transfer from donor to acceptor cells are extremely important. Here, we summarize the methods EV are labeled and their functional transfer in acceptor cells are evaluated by various reporter systems.
Collapse
Affiliation(s)
- Chaoshan Han
- Department of Biomedical Engineering, School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
- Department of Biomedical Engineering, School of Engineering, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Gangjian Qin
- Department of Biomedical Engineering, School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
- Department of Biomedical Engineering, School of Engineering, University of Alabama at Birmingham, Birmingham, AL, United States
- *Correspondence: Gangjian Qin,
| |
Collapse
|
91
|
Xie Y, Guan Q, Guo J, Chen Y, Yin Y, Han X. Hydrogels for Exosome Delivery in Biomedical Applications. Gels 2022; 8:gels8060328. [PMID: 35735672 PMCID: PMC9223116 DOI: 10.3390/gels8060328] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 05/20/2022] [Accepted: 05/21/2022] [Indexed: 02/08/2023] Open
Abstract
Hydrogels, which are hydrophilic polymer networks, have attracted great attention, and significant advances in their biological and biomedical applications, such as for drug delivery, tissue engineering, and models for medical studies, have been made. Due to their similarity in physiological structure, hydrogels are highly compatible with extracellular matrices and biological tissues and can be used as both carriers and matrices to encapsulate cellular secretions. As small extracellular vesicles secreted by nearly all mammalian cells to mediate cell–cell interactions, exosomes play very important roles in therapeutic approaches and disease diagnosis. To maintain their biological activity and achieve controlled release, a strategy that embeds exosomes in hydrogels as a composite system has been focused on in recent studies. Therefore, this review aims to provide a thorough overview of the use of composite hydrogels for embedding exosomes in medical applications, including the resources for making hydrogels and the properties of hydrogels, and strategies for their combination with exosomes.
Collapse
Affiliation(s)
- Yaxin Xie
- State Key Laboratory of Oral Diseases, Department of Orthodontics, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China; (Y.X.); (J.G.); (Y.C.); (Y.Y.)
| | - Qiuyue Guan
- Department of Geriatrics, People’s Hospital of Sichuan Province, Chengdu 610041, China;
| | - Jiusi Guo
- State Key Laboratory of Oral Diseases, Department of Orthodontics, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China; (Y.X.); (J.G.); (Y.C.); (Y.Y.)
| | - Yilin Chen
- State Key Laboratory of Oral Diseases, Department of Orthodontics, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China; (Y.X.); (J.G.); (Y.C.); (Y.Y.)
| | - Yijia Yin
- State Key Laboratory of Oral Diseases, Department of Orthodontics, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China; (Y.X.); (J.G.); (Y.C.); (Y.Y.)
| | - Xianglong Han
- State Key Laboratory of Oral Diseases, Department of Orthodontics, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China; (Y.X.); (J.G.); (Y.C.); (Y.Y.)
- Correspondence:
| |
Collapse
|
92
|
Gunnels TF, Stranford DM, Mitrut RE, Kamat NP, Leonard JN. Elucidating Design Principles for Engineering Cell-Derived Vesicles to Inhibit SARS-CoV-2 Infection. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2200125. [PMID: 35388947 PMCID: PMC9106922 DOI: 10.1002/smll.202200125] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 02/21/2022] [Indexed: 06/14/2023]
Abstract
The ability of pathogens to develop drug resistance is a global health challenge. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) presents an urgent need wherein several variants of concern resist neutralization by monoclonal antibody (mAb) therapies and vaccine-induced sera. Decoy nanoparticles-cell-mimicking particles that bind and inhibit virions-are an emerging class of therapeutics that may overcome such drug resistance challenges. To date, quantitative understanding as to how design features impact performance of these therapeutics is lacking. To address this gap, this study presents a systematic, comparative evaluation of various biologically derived nanoscale vesicles, which may be particularly well suited to sustained or repeated administration in the clinic due to low toxicity, and investigates their potential to inhibit multiple classes of model SARS-CoV-2 virions. A key finding is that such particles exhibit potent antiviral efficacy across multiple manufacturing methods, vesicle subclasses, and virus-decoy binding affinities. In addition, these cell-mimicking vesicles effectively inhibit model SARS-CoV-2 variants that evade mAbs and recombinant protein-based decoy inhibitors. This study provides a foundation of knowledge that may guide the design of decoy nanoparticle inhibitors for SARS-CoV-2 and other viral infections.
Collapse
Affiliation(s)
- Taylor F. Gunnels
- Department of Biomedical EngineeringNorthwestern UniversityEvanstonIL60208USA
- Center for Synthetic BiologyNorthwestern UniversityEvanstonIL60208USA
| | - Devin M. Stranford
- Center for Synthetic BiologyNorthwestern UniversityEvanstonIL60208USA
- Department of Chemical and Biological EngineeringNorthwestern UniversityEvanstonIL60208USA
| | - Roxana E. Mitrut
- Center for Synthetic BiologyNorthwestern UniversityEvanstonIL60208USA
- Department of Chemical and Biological EngineeringNorthwestern UniversityEvanstonIL60208USA
| | - Neha P. Kamat
- Department of Biomedical EngineeringNorthwestern UniversityEvanstonIL60208USA
- Center for Synthetic BiologyNorthwestern UniversityEvanstonIL60208USA
- Chemistry of Life Processes InstituteNorthwestern UniversityEvanstonIL60208USA
| | - Joshua N. Leonard
- Center for Synthetic BiologyNorthwestern UniversityEvanstonIL60208USA
- Department of Chemical and Biological EngineeringNorthwestern UniversityEvanstonIL60208USA
- Chemistry of Life Processes InstituteNorthwestern UniversityEvanstonIL60208USA
- Robert H. Lurie Comprehensive Cancer CenterNorthwestern UniversityEvanstonIL60208USA
| |
Collapse
|
93
|
Durán-Jara E, Vera-Tobar T, Lobos-González LDL. Lactadherin: From a Well-Known Breast Tumor Marker to a Possible Player in Extracellular Vesicle-Mediated Cancer Progression. Int J Mol Sci 2022; 23:3855. [PMID: 35409215 PMCID: PMC8998968 DOI: 10.3390/ijms23073855] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 03/25/2022] [Accepted: 03/26/2022] [Indexed: 02/04/2023] Open
Abstract
Lactadherin is a secreted glycoprotein associated with the milk fat globule membrane, which is highly present in the blood and in the mammary tissue of lactating women. Several biological functions have been associated with this protein, mainly attributable to its immunomodulatory role promoting phagocyte-mediated clearance of apoptotic cells. It has been shown that lactadherin also plays important roles in cell adhesion, the promotion of angiogenesis, and tissue regeneration. On the other hand, this protein has been used as a marker of breast cancer and tumor progression. Recently, high levels of lactadherin has been associated with poor prognosis and decreased survival, not only in breast cancer, but also in melanoma, ovarian, colorectal, and other types of cancer. Although the mechanisms responsible for the tumor-promoting effects attributed to lactadherin have not been fully elucidated, a growing body of literature indicates that lactadherin could be a promising therapeutic target and/or biomarker for breast and other tumors. Moreover, recent studies have shown its presence in extracellular vesicles derived from cancer cell lines and cancer patients, which was associated with cancer aggressiveness and worse prognosis. Thus, this review will focus on the link between lactadherin and cancer development and progression, its possible use as a cancer biomarker and/or therapeutic target, concluding with a possible role of this protein in cellular communication mediated by extracellular vesicles.
Collapse
Affiliation(s)
- Eduardo Durán-Jara
- Centro de Medicina Regenerativa, Instituto de Ciencias e Innovación en Medicina, Facultad de Medicina Clínica Alemana, Universidad del Desarrollo, Santiago 7710162, Chile; (E.D.-J.); (T.V.-T.)
| | - Tamara Vera-Tobar
- Centro de Medicina Regenerativa, Instituto de Ciencias e Innovación en Medicina, Facultad de Medicina Clínica Alemana, Universidad del Desarrollo, Santiago 7710162, Chile; (E.D.-J.); (T.V.-T.)
| | - Lorena De Lourdes Lobos-González
- Centro de Medicina Regenerativa, Instituto de Ciencias e Innovación en Medicina, Facultad de Medicina Clínica Alemana, Universidad del Desarrollo, Santiago 7710162, Chile; (E.D.-J.); (T.V.-T.)
- Advanced Center for Chronic Diseases (ACCDiS), Independencia, Santiago 8380000, Chile
| |
Collapse
|
94
|
Bost JP, Saher O, Hagey D, Mamand DR, Liang X, Zheng W, Corso G, Gustafsson O, Görgens A, Smith CIE, Zain R, El Andaloussi S, Gupta D. Growth Media Conditions Influence the Secretion Route and Release Levels of Engineered Extracellular Vesicles. Adv Healthc Mater 2022; 11:e2101658. [PMID: 34773385 PMCID: PMC11469210 DOI: 10.1002/adhm.202101658] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 11/01/2021] [Indexed: 12/12/2022]
Abstract
Extracellular vesicles (EVs) are nanosized cell-derived vesicles produced by all cells, which provide a route of intercellular communication by transmitting biological cargo. While EVs offer promise as therapeutic agents, the molecular mechanisms of EV biogenesis are not yet fully elucidated, in part due to the concurrence of numerous interwoven pathways which give rise to heterogenous EV populations in vitro. The equilibrium between the EV-producing pathways is heavily influenced by factors in the extracellular environment, in such a way that can be taken advantage of to boost production of engineered EVs. In this study, a quantifiable EV-engineering approach is used to investigate how different cell media conditions alter EV production. The presence of serum, exogenous EVs, and other signaling factors in cell media alters EV production at the physical, molecular, and transcriptional levels. Further, it is demonstrated that the ceramide-dependent EV biogenesis route is the major pathway to production of engineered EVs during optimized EV-production. These findings suggest a novel understanding to the mechanisms underlying EV production in cell culture which can be applied to develop advanced EV production methods.
Collapse
Affiliation(s)
- Jeremy P. Bost
- Department of Laboratory MedicineKarolinska InstitutetHuddinge14152Sweden
| | - Osama Saher
- Department of Laboratory MedicineKarolinska InstitutetHuddinge14152Sweden
- Department of Pharmaceutics and Industrial PharmacyFaculty of PharmacyCairo UniversityCairo11562Egypt
| | - Daniel Hagey
- Department of Laboratory MedicineKarolinska InstitutetHuddinge14152Sweden
| | - Doste R. Mamand
- Department of Laboratory MedicineKarolinska InstitutetHuddinge14152Sweden
- Department of BiologyFaculty of ScienceCihan University‐ErbilArbil5XC8+WVIraq
| | - Xiuming Liang
- Department of Laboratory MedicineKarolinska InstitutetHuddinge14152Sweden
| | - Wenyi Zheng
- Department of Laboratory MedicineKarolinska InstitutetHuddinge14152Sweden
| | - Giulia Corso
- Department of Laboratory MedicineKarolinska InstitutetHuddinge14152Sweden
| | - Oskar Gustafsson
- Department of Laboratory MedicineKarolinska InstitutetHuddinge14152Sweden
| | - André Görgens
- Department of Laboratory MedicineKarolinska InstitutetHuddinge14152Sweden
| | - CI Edvard Smith
- Department of Laboratory MedicineKarolinska InstitutetHuddinge14152Sweden
| | - Rula Zain
- Department of Laboratory MedicineKarolinska InstitutetHuddinge14152Sweden
- Centre for Rare DiseasesDepartment of Clinical GeneticsKarolinska University HospitalStockholmSE‐171 76Sweden
| | | | - Dhanu Gupta
- Department of Laboratory MedicineKarolinska InstitutetHuddinge14152Sweden
| |
Collapse
|
95
|
Fan J, Pan J, Zhang X, Chen Y, Zeng Y, Huang L, Ma D, Chen Z, Wu G, Fan W. A peptide derived from the N-terminus of charged multivesicular body protein 6 (CHMP6) promotes the secretion of gene editing proteins via small extracellular vesicle production. Bioengineered 2022; 13:4702-4716. [PMID: 35188876 PMCID: PMC8973635 DOI: 10.1080/21655979.2022.2030571] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Extracellular vesicles (EVs) are a promising new therapeutic platform. However, the low cargo-loading efficiency limits their clinical translation. In this study, we developed a high-yield EV cargo-loading device and explored its ability to encapsulate gene editing proteins. A series of fusion protein-based systems were constructed and their cargo loading efficiencies were compared by a NanoGlo luciferase assay. A myristoylated (Myr) peptide tag cloned from the N-terminal region of charged multivesicular body protein 6 (CHMP6), termed Myr(CHMP6), outcompeted CD9, ARRDC1, and other short polypeptides as an active packaging device. As determined by nanoparticle tracking analysis and transmission electron microscopy, the overexpression of Myr(CHMP6) increased small EV (sEV) production in Lenti-X 293T cells without altering sEV morphology. The high passive packaging efficiency of Myr(CHMP6) was also elucidated for unmodified cargo loading. Western blotting revealed that Myr(CHMP6) facilitated the loading of Cre and Cas9 into sEVs without the generation of packaging device-cargo fusion proteins. Furthermore, Myr(CHMP6)-modified sEVs loaded with Cre or Cas9 promoted gene-editing in recipient cells, as observed using a fluorescence reporter system. Subsequent investigation demonstrated a dose-dependent effect of Myr(CHMP6) tag-induced cargo-loading. Mechanistically, N-myristoylation alone was necessary but not sufficient for the effective packaging of proteins into EVs. Thus, our results indicated that Myr(CHMP6) induces sEV production and may be effective in loading gene editing proteins into sEVs for therapeutic purposes.
Collapse
Affiliation(s)
- Junyu Fan
- Department of Cardiology The Eighth Affiliated Hospital of Sun Yat-sen University Guangdong Shenzhen P.R. China.,NHC Key Laboratory of Assisted Circulation, Sun Yat-Sen University, Guangdong, Guangzhou, P.R. China
| | - Jiajie Pan
- Department of Cardiology The Eighth Affiliated Hospital of Sun Yat-sen University Guangdong Shenzhen P.R. China.,NHC Key Laboratory of Assisted Circulation, Sun Yat-Sen University, Guangdong, Guangzhou, P.R. China
| | - Xiaozhe Zhang
- Department of Cardiology The Eighth Affiliated Hospital of Sun Yat-sen University Guangdong Shenzhen P.R. China.,NHC Key Laboratory of Assisted Circulation, Sun Yat-Sen University, Guangdong, Guangzhou, P.R. China
| | - Yixi Chen
- Department of Cardiology The Eighth Affiliated Hospital of Sun Yat-sen University Guangdong Shenzhen P.R. China.,NHC Key Laboratory of Assisted Circulation, Sun Yat-Sen University, Guangdong, Guangzhou, P.R. China
| | - Yue Zeng
- Department of Cardiology The Eighth Affiliated Hospital of Sun Yat-sen University Guangdong Shenzhen P.R. China.,NHC Key Laboratory of Assisted Circulation, Sun Yat-Sen University, Guangdong, Guangzhou, P.R. China
| | - Lihan Huang
- Department of Cardiology The Eighth Affiliated Hospital of Sun Yat-sen University Guangdong Shenzhen P.R. China.,NHC Key Laboratory of Assisted Circulation, Sun Yat-Sen University, Guangdong, Guangzhou, P.R. China
| | - Dongwei Ma
- Department of Cardiology The Eighth Affiliated Hospital of Sun Yat-sen University Guangdong Shenzhen P.R. China.,NHC Key Laboratory of Assisted Circulation, Sun Yat-Sen University, Guangdong, Guangzhou, P.R. China
| | - Ziqi Chen
- Department of Cardiology The Eighth Affiliated Hospital of Sun Yat-sen University Guangdong Shenzhen P.R. China.,NHC Key Laboratory of Assisted Circulation, Sun Yat-Sen University, Guangdong, Guangzhou, P.R. China
| | - Guifu Wu
- Department of Cardiology The Eighth Affiliated Hospital of Sun Yat-sen University Guangdong Shenzhen P.R. China.,NHC Key Laboratory of Assisted Circulation, Sun Yat-Sen University, Guangdong, Guangzhou, P.R. China.,Guangdong Innovative Engineering and Technology Research Center for Assisted Circulation Guangdong Shenzhen P.R. China
| | - Wendong Fan
- Department of Cardiology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P.R. China.,NHC Key Laboratory of Assisted Circulation, Sun Yat-Sen University, Guangdong, Guangzhou, P.R. China
| |
Collapse
|
96
|
Wang BZ, Luo L, Vunjak-Novakovic G. RNA and Protein Delivery by Cell-Secreted and Bioengineered Extracellular Vesicles. Adv Healthc Mater 2022; 11:e2101557. [PMID: 34706168 PMCID: PMC8891029 DOI: 10.1002/adhm.202101557] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 09/28/2021] [Indexed: 12/22/2022]
Abstract
Extracellular vesicles (EVs) are carriers of biological signals through export and delivery of RNAs and proteins. Of increasing interest is the use of EVs as a platform for delivery of biomolecules. Preclinical studies have effectively used EVs to treat a number of diseases. Uniquely, endogenous machinery within cells can be manipulated in order to produce desirable loading of cargo within secreted EVs. In order to inform the development of such approaches, an understanding of the cellular mechanisms by which cargo is sorted to EVs is required. Here, the current knowledge of cargo sorting within EVs is reviewed. Here is given an overview of recent bioengineering approaches that leverage these advances. Methods of externally manipulating EV cargo are also discussed. Finally, a perspective on the current challenges of EVs as a drug delivery platform is offered. It is proposed that standardized bioengineering methods for therapeutic EV preparation will be required to create a well-defined clinical product.
Collapse
Affiliation(s)
- Bryan Z. Wang
- Department of Biomedical Engineering, 622 West 168th Street VC12-234, 10032, U.S.A
- Department of Medicine, 622 West 168th Street VC12-234, 10032, U.S.A
| | - Lori Luo
- Department of Medicine, 622 West 168th Street VC12-234, 10032, U.S.A
| | - Gordana Vunjak-Novakovic
- Department of Biomedical Engineering, 622 West 168th Street VC12-234, 10032, U.S.A
- Department of Medicine, 622 West 168th Street VC12-234, 10032, U.S.A
| |
Collapse
|
97
|
Xu M, Chen G, Dong Y, Yang J, Liu Y, Song H, Song H, Wang Y. Liraglutide Loaded-Milk Exosomes Lower Blood Glucose when Given by Sublingual Route. ChemMedChem 2022; 17:e202100758. [PMID: 35199481 DOI: 10.1002/cmdc.202100758] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 02/23/2022] [Indexed: 11/09/2022]
Abstract
Bovine milk is rich in extracellular vesicles (mEVs) which have been suggested as a possible drug delivery vehicle with oral bioavailability. As the digestive fluids contain many lipid- and protein-degrading enzymes, we explored whether mEVs given sublingually could be taken up systemically. mEVs were isolated using three different protocols, which were 120 nm in diameter and carried bovine CD81. Fluorescently stained mEVs given by sublingual route were detected in the circulation, whereas mEVs given by gavage were detected at 2-Log lower concentrations. As proof of the concept, we loaded mEVs with the antidiabetic drug Liraglutide (LRT-EV), which reduced blood glucose levels when given by the sublingual route but showed no efficacy via gavaging. This study suggests that mEV may be an efficient delivery vehicle for drugs that are not orally bioavailable, and LRT-loaded EVs have the potential as the next-generation drug delivery platform for the treatment of chronic diseases, including diabetes.
Collapse
Affiliation(s)
- Mingzhi Xu
- National Center of Protein Sciences Beijing, Beijing Institute of Lifeomics, 37 ZGC Life Sciences Park Rd., Changping District, Beijing, CHINA
| | - Gang Chen
- National Center of Protein Sciences Beijing, Beijing Institute of Lifeomics, 37 ZGC Life Sciences Park Rd., Changping District, Beijing, CHINA
| | - Yanan Dong
- National Center of Protein Sciences Beijing, Beijing Institute of Lifeomics, 37 ZGC Life Sciences Park Rd., Changping District, Beijing, CHINA
| | - Jing Yang
- National Center of Protein Sciences Beijing, Beijing Institute of Lifeomics, 37 ZGC Life Sciences Park Rd., Changping District, Beijing, CHINA
| | - Yongxue Liu
- Beijing Institute of Radiation Medicine, Pharmacology, 27 Bei Taiping Rd., Haidian District, Beijing, CHINA
| | - Haijing Song
- PLA Strategic Support Force Medical Center, Emergency Medicine, PLA Strategic Support Force Medical Center, Anxiang Bei Rd., Chaoyang District, Beijing, CHINA
| | - Haifeng Song
- National Center of Protein Sciences Beijing, Beijing Institute of Lifeomics, 37 ZGC Life Sciences Park Rd., Changping District, Beijing, CHINA
| | - Yi Wang
- National Center of Protein Sciences Beijing, Beijing Institute of Lifeomics, 37 ZGC Life Sciences Park Rd., Changping District, 102206, Beijing, CHINA
| |
Collapse
|
98
|
Zarubova J, Hasani‐Sadrabadi MM, Dashtimoghadam E, Zhang X, Ansari S, Li S, Moshaverinia A. Engineered Delivery of Dental Stem-Cell-Derived Extracellular Vesicles for Periodontal Tissue Regeneration. Adv Healthc Mater 2022; 11:e2102593. [PMID: 35191610 DOI: 10.1002/adhm.202102593] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/31/2022] [Indexed: 11/05/2022]
Abstract
Periodontal disease begins as an inflammatory response to a bacterial biofilm deposited around the teeth, which over time leads to the destruction of tooth-supporting structures and consequently tooth loss. Conventional treatment strategies show limited efficacy in promoting regeneration of damaged periodontal tissues. Here, a delivery platform is developed for small extracellular vesicles (sEVs) derived from gingival mesenchymal stem cells (GMSCs) to treat periodontitis. EVs can achieve comparable therapeutic effects to their cells of origin. However, the short half-lives of EVs after their administration along with their rapid diffusion away from the delivery site necessitate frequent administration to achieve therapeutic benefits. To address these issues, "dual delivery" microparticles are engineered enabling microenvironment-sensitive release of EVs by metalloproteinases at the affected site along with antibiotics to suppress bacterial biofilm growth. GMSC sEVs are able to decrease the secretion of pro-inflammatory cytokines by monocytes/macrophages and T cells, suppress T-cell activation, and induce the formation of T regulatory cells (Tregs) in vitro and in a rat model of periodontal disease. One-time administration of immunomodulatory GMSC sEV-decorated microparticles leads to a significant improvement in regeneration of the damaged periodontal tissue. This approach will have potential clinical applications in the regeneration of a variety of tissues.
Collapse
Affiliation(s)
- Jana Zarubova
- Department of Bioengineering University of California 420 Westwood Plaza, 5121 Engineering V Los Angeles CA 90095‐1600 USA
- Department of Biomaterials and Tissue Engineering Institute of Physiology of the Czech Academy of Sciences Prague 14220 Czech Republic
| | | | - Erfan Dashtimoghadam
- Department of Chemistry University of North Carolina at Chapel Hill Chapel Hill NC 27599‐3290 USA
| | - Xuexiang Zhang
- Department of Bioengineering University of California 420 Westwood Plaza, 5121 Engineering V Los Angeles CA 90095‐1600 USA
| | - Sahar Ansari
- Weintraub Center for Reconstructive Biotechnology Division of Advanced Prosthodontics School of Dentistry University of California Los Angeles CA 90095 USA
| | - Song Li
- Department of Bioengineering University of California 420 Westwood Plaza, 5121 Engineering V Los Angeles CA 90095‐1600 USA
| | - Alireza Moshaverinia
- Weintraub Center for Reconstructive Biotechnology Division of Advanced Prosthodontics School of Dentistry University of California Los Angeles CA 90095 USA
| |
Collapse
|
99
|
Kanada M, Linenfelser L, Cox E, Gilad AA. A Dual-Reporter Platform for Screening Tumor-Targeted Extracellular Vesicles. Pharmaceutics 2022; 14:475. [PMID: 35335849 PMCID: PMC8953635 DOI: 10.3390/pharmaceutics14030475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 02/10/2022] [Accepted: 02/18/2022] [Indexed: 12/10/2022] Open
Abstract
Extracellular vesicle (EV)-mediated transfer of biomolecules plays an essential role in intercellular communication and may improve targeted drug delivery. In the past decade, various approaches to EV surface modification for targeting specific cells or tissues have been proposed, including genetic engineering of parental cells or postproduction EV engineering. However, due to technical limitations, targeting moieties of engineered EVs have not been thoroughly characterized. Here, we report the bioluminescence resonance energy transfer (BRET) EV reporter, PalmReNL-based dual-reporter platform for characterizing the cellular uptake of tumor-homing peptide (THP)-engineered EVs, targeting PDL1, uPAR, or EGFR proteins expressed in MDA-MB-231 breast cancer cells, simultaneously by bioluminescence measurement and fluorescence microscopy. Bioluminescence analysis of cellular EV uptake revealed the highest binding efficiency of uPAR-targeted EVs, whereas PDL1-targeted EVs showed slower cellular uptake. EVs engineered with two known EGFR-binding peptides via lipid nanoprobes did not increase cellular uptake, indicating that designs of EGFR-binding peptide conjugation to the EV surface are critical for functional EV engineering. Fluorescence analysis of cellular EV uptake allowed us to track individual PalmReNL-EVs bearing THPs in recipient cells. These results demonstrate that the PalmReNL-based EV assay platform can be a foundation for high-throughput screening of tumor-targeted EVs.
Collapse
Affiliation(s)
- Masamitsu Kanada
- Institute for Quantitative Health Science and Engineering (IQ), Michigan State University, East Lansing, MI 48824, USA; (L.L.); (E.C.)
- Department of Pharmacology & Toxicology, Michigan State University, East Lansing, MI 48824, USA
| | - Lauren Linenfelser
- Institute for Quantitative Health Science and Engineering (IQ), Michigan State University, East Lansing, MI 48824, USA; (L.L.); (E.C.)
- Department of Chemical Engineering & Materials Science, Michigan State University, East Lansing, MI 48824, USA
| | - Elyssa Cox
- Institute for Quantitative Health Science and Engineering (IQ), Michigan State University, East Lansing, MI 48824, USA; (L.L.); (E.C.)
| | - Assaf A. Gilad
- Institute for Quantitative Health Science and Engineering (IQ), Michigan State University, East Lansing, MI 48824, USA; (L.L.); (E.C.)
- Department of Chemical Engineering & Materials Science, Michigan State University, East Lansing, MI 48824, USA
- Department of Radiology, Michigan State University, East Lansing, MI 48824, USA
| |
Collapse
|
100
|
Thomas SC, Kim JW, Pauletti GM, Hassett DJ, Kotagiri N. Exosomes: Biological Pharmaceutical Nanovectors for Theranostics. Front Bioeng Biotechnol 2022; 9:808614. [PMID: 35096795 PMCID: PMC8790084 DOI: 10.3389/fbioe.2021.808614] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 12/24/2021] [Indexed: 12/12/2022] Open
Abstract
Exosomes are natural cell-derived nanovesicles of endocytic origin that enable cellular crosstalk by transferring encapsulated molecular cargos across biological barriers, thereby holding significantly complex implications in the etiology and progression of diverse disease states. Consequently, the development of exosomes-based nano-theranostic strategies has received immense consideration for advancing therapeutic interventions and disease prognosis. Their favorable biopharmaceutical properties make exosomes a unique nanoparticulate carrier for pharmaceutical drug delivery. This review provides an update on the contemporary strategies utilizing exosomes for theranostic applications in nanomedicine. In addition, we provide a synopsis of exosomal features and insights into strategic modifications that control in vivo biodistribution. We further discuss their opportunities, merits and pitfalls for cell/tissue targeted drug delivery in personalized nanotherapy.
Collapse
Affiliation(s)
- Shindu C Thomas
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, OH, United States
| | - Jin-Woo Kim
- Department of Biological and Agricultural Engineering, Institute for Nanoscience and Engineering, University of Arkansas, Fayetteville, AR, United States
| | - Giovanni M Pauletti
- St. Louis College of Pharmacy, University of Health Sciences and Pharmacy in St. Louis, St. Louis, MO, United States
| | - Daniel J Hassett
- Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Nalinikanth Kotagiri
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, OH, United States
| |
Collapse
|