1
|
Offens A, Teeuwen L, Gucluler Akpinar G, Steiner L, Kooijmans S, Mamand D, Weissinger H, Käll A, Eldh M, Wiklander OPB, El-Andaloussi S, Karlsson MCI, Vader P, Gabrielsson S. A fusion protein that targets antigen-loaded extracellular vesicles to B cells enhances antigen-specific T cell expansion. J Control Release 2025; 382:113665. [PMID: 40147536 DOI: 10.1016/j.jconrel.2025.113665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Revised: 03/15/2025] [Accepted: 03/24/2025] [Indexed: 03/29/2025]
Abstract
Extracellular vesicles (EVs) have the potential to modulate immune responses via their cargo molecules and are being explored as vehicles in cancer immunotherapy. Dendritic cell-derived EVs can induce antigen-specific immune responses leading to reduced tumor burden. This response was shown to depend partially on B cells. EVs can be targeted to certain cells or tissues, and EVs from Epstein-Barr Virus (EBV) infected cells were shown to carry the EBV glycoprotein GP350 on their surface and target human CD21 (hCD21) on B cells. We therefore investigated whether targeting EVs to B cells via this mechanism could improve antigen-specific immune responses. A soluble fusion protein containing the phosphatidylserine-binding domain (C1C2) of lactadherin and hCD21-binding domain (D123) of GP350 was used to decorate and target EVs to B cells. D123-decorated EVs increased in vitro B cell targeting 5-fold compared to EVs decorated with a non-targeting control protein or undecorated EVs. Furthermore, in vivo, D123-decoration did not alter the biodistribution of EVs across organs but specifically targeted them to B cells in the spleen, blood and lymph nodes of hCD21-transgenic mice. Immunization with hCD21-targeted, OVA-loaded EVs resulted in a higher percentage of antigen-specific CD8+ T cells compared to untargeted EVs. Our data show that D123-decorated EVs efficiently target B cells and improve antigen-specific T cell responses in vivo, which could be explored in future therapeutic applications.
Collapse
Affiliation(s)
- Annemarijn Offens
- Division of Immunology and Respiratory Medicine, Department of Medicine, Karolinska Institutet, Center for Molecular Medicine (CMM L8:00), Visionsgatan 18, 171 64 Solna, Sweden; Department of Clinical Immunology and Transfusion Medicine, Karolinska University Hospital, Stockholm, Sweden; Center for Molecular Medicine (CMM), Stockholm, Sweden
| | - Loes Teeuwen
- Division of Immunology and Respiratory Medicine, Department of Medicine, Karolinska Institutet, Center for Molecular Medicine (CMM L8:00), Visionsgatan 18, 171 64 Solna, Sweden; Department of Clinical Immunology and Transfusion Medicine, Karolinska University Hospital, Stockholm, Sweden; Center for Molecular Medicine (CMM), Stockholm, Sweden
| | - Gozde Gucluler Akpinar
- Division of Immunology and Respiratory Medicine, Department of Medicine, Karolinska Institutet, Center for Molecular Medicine (CMM L8:00), Visionsgatan 18, 171 64 Solna, Sweden; Department of Clinical Immunology and Transfusion Medicine, Karolinska University Hospital, Stockholm, Sweden; Center for Molecular Medicine (CMM), Stockholm, Sweden
| | - Loïc Steiner
- Division of Immunology and Respiratory Medicine, Department of Medicine, Karolinska Institutet, Center for Molecular Medicine (CMM L8:00), Visionsgatan 18, 171 64 Solna, Sweden; Department of Clinical Immunology and Transfusion Medicine, Karolinska University Hospital, Stockholm, Sweden; Center for Molecular Medicine (CMM), Stockholm, Sweden
| | - Sander Kooijmans
- CDL Research, University Medical Center, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands; Metabolic Diseases, Wilhelmina's Children's Hospital, University Medical Center Utrecht, Utrecht, the Netherlands; Regenerative Medicine Center, Uppsalalaan 8, 3584 CT Utrecht, The Netherlands
| | - Doste Mamand
- Division of Biomolecular and Cellular Medicine, Department of Laboratory Medicine, Karolinska Institutet, ANA Futura, Alfred-Nobels-Allé 8, Huddinge, Stockholm 14152, Sweden; Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST), Karolinska University Hospital, Stockholm, Sweden; Karolinska ATMP Center, Karolinska Institutet, Huddinge, Stockholm, Sweden; Breast Center, Karolinska Comprehensive Cancer Center, Karolinska University Hospital, Stockholm, Sweden
| | - Hannah Weissinger
- Division of Immunology and Respiratory Medicine, Department of Medicine, Karolinska Institutet, Center for Molecular Medicine (CMM L8:00), Visionsgatan 18, 171 64 Solna, Sweden; Department of Clinical Immunology and Transfusion Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Alexander Käll
- Division of Immunology and Respiratory Medicine, Department of Medicine, Karolinska Institutet, Center for Molecular Medicine (CMM L8:00), Visionsgatan 18, 171 64 Solna, Sweden; Department of Clinical Immunology and Transfusion Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Maria Eldh
- Division of Immunology and Respiratory Medicine, Department of Medicine, Karolinska Institutet, Center for Molecular Medicine (CMM L8:00), Visionsgatan 18, 171 64 Solna, Sweden; Department of Clinical Immunology and Transfusion Medicine, Karolinska University Hospital, Stockholm, Sweden; Center for Molecular Medicine (CMM), Stockholm, Sweden
| | - Oscar P B Wiklander
- Division of Biomolecular and Cellular Medicine, Department of Laboratory Medicine, Karolinska Institutet, ANA Futura, Alfred-Nobels-Allé 8, Huddinge, Stockholm 14152, Sweden; Karolinska ATMP Center, Karolinska Institutet, Huddinge, Stockholm, Sweden; Breast Center, Karolinska Comprehensive Cancer Center, Karolinska University Hospital, Stockholm, Sweden
| | - Samir El-Andaloussi
- Division of Biomolecular and Cellular Medicine, Department of Laboratory Medicine, Karolinska Institutet, ANA Futura, Alfred-Nobels-Allé 8, Huddinge, Stockholm 14152, Sweden; Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST), Karolinska University Hospital, Stockholm, Sweden; Karolinska ATMP Center, Karolinska Institutet, Huddinge, Stockholm, Sweden
| | - Mikael C I Karlsson
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Biomedicum, Solnavägen 9, C7, 17165 Solna, Sweden
| | - Pieter Vader
- CDL Research, University Medical Center, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
| | - Susanne Gabrielsson
- Division of Immunology and Respiratory Medicine, Department of Medicine, Karolinska Institutet, Center for Molecular Medicine (CMM L8:00), Visionsgatan 18, 171 64 Solna, Sweden; Department of Clinical Immunology and Transfusion Medicine, Karolinska University Hospital, Stockholm, Sweden; Center for Molecular Medicine (CMM), Stockholm, Sweden.
| |
Collapse
|
2
|
Hermann DM, Wang C, Mohamud Yusuf A, Herz J, Doeppner TR, Giebel B. Extracellular vesicles lay the ground for neuronal plasticity by restoring mitochondrial function, cell metabolism and immune balance. J Cereb Blood Flow Metab 2025:271678X251325039. [PMID: 40072028 PMCID: PMC11904928 DOI: 10.1177/0271678x251325039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Revised: 01/15/2025] [Accepted: 02/14/2025] [Indexed: 03/15/2025]
Abstract
Extracellular vesicles (EVs) convey complex signals between cells that can be used to promote neuronal plasticity and neurological recovery in brain disease models. These EV signals are multimodal and context-dependent, making them unique therapeutic principles. This review analyzes how EVs released from various cell sources control neuronal metabolic function, neuronal survival and plasticity. Preferential sites of EV communication in the brain are interfaces between pre- and postsynaptic neurons at synapses, between astrocytes and neurons at plasma membranes or tripartite synapses, between oligodendrocytes and neurons at axons, between microglial cells/macrophages and neurons, and between cerebral microvascular cells and neurons. At each of these interfaces, EVs support mitochondrial function and cell metabolism under physiological conditions and orchestrate neuronal survival and plasticity in response to brain injury. In the injured brain, the promotion of neuronal survival and plasticity by EVs is tightly linked with EV actions on mitochondrial function, cell metabolism, oxidative stress and immune responses. Via the stabilization of cell metabolism and immune balance, neuronal plasticity responses are activated and functional neurological recovery is induced. As such, EV lay the ground for neuronal plasticity.
Collapse
Affiliation(s)
- Dirk M Hermann
- Department of Neurology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Chen Wang
- Department of Neurology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Ayan Mohamud Yusuf
- Department of Neurology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Josephine Herz
- Department of Pediatrics I, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Thorsten R Doeppner
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Bernd Giebel
- Department of Neurology, University Hospital Gießen and Marburg, Justus-Liebig-University Gießen, Gießen, Germany
| |
Collapse
|
3
|
Simon L, Constanzo J, Terraza-Aguirre C, Ibn Elfekih Z, Berthelot J, Benkhaled BT, Haute T, Pednekar K, Clark K, Emerson SJ, Atis S, Benedetti C, Langlois S, Marquant A, Prakash J, Wang A, Devoisselle JM, Montier T, Djouad F, Pouget JP, Lapinte V, Morille M. Surface modification of extracellular vesicles with polyoxazolines to enhance their plasma stability and tumor accumulation. Biomaterials 2025; 313:122748. [PMID: 39180918 DOI: 10.1016/j.biomaterials.2024.122748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 07/23/2024] [Accepted: 08/05/2024] [Indexed: 08/27/2024]
Abstract
Extracellular vesicles (EVs) are future promising therapeutics, but their instability in vivo after administration remains an important barrier to their further development. Many groups evaluated EV surface modification strategies to add a targeting group with the aim of controlling EV biodistribution. Conversely, fewer groups focused on their stabilization to obtain "stealth" allogenic EVs. Modulating their stabilization and biodistribution is an essential prerequisite for their development as nano-therapeutics. Here, we explored polyoxazolines with lipid anchors association to the EV membrane (POxylation as an alternative to PEGylation) to stabilize EVs in plasma and control their biodistribution, while preserving their native properties. We found that this modification maintained and seemed to potentiate the immunomodulatory properties of EVs derived from mesenchymal stem/stromal cells (MSC). Using a radiolabeling protocol to track EVs at a therapeutically relevant concentration in vivo, we demonstrated that POxylation is a promising option to stabilize EVs in plasma because it increased EV half-life by 6 fold at 6 h post-injection. Moreover, EV accumulation in tumors was higher after POxylation than after PEGylation.
Collapse
Affiliation(s)
- L Simon
- ICGM, Montpellier University, CNRS, ENSCM, Montpellier, France
| | - J Constanzo
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, Université de Montpellier, Institut Régional Du Cancer de Montpellier (ICM), Montpellier, France
| | | | - Z Ibn Elfekih
- ICGM, Montpellier University, CNRS, ENSCM, Montpellier, France
| | - J Berthelot
- ICGM, Montpellier University, CNRS, ENSCM, Montpellier, France
| | - B T Benkhaled
- ICGM, Montpellier University, CNRS, ENSCM, Montpellier, France
| | - T Haute
- Univ Brest, INSERM, EFS, UMR 1078, GGB, F-29200, Brest, France
| | - K Pednekar
- Department of Advanced Organ Bioengineering and Therapeutics, Engineered Therapeutics Section, Technical Medical Centre, University of Twente, 7500 AE, Enschede, the Netherlands
| | - K Clark
- Center for Surgical Bioengineering, Deparment of Surgery, University of California Davis School of Medicine, Sacramento, CA, USA; Institute for Pediatric Regenerative Medicine, Shriners Children's Northern California, Sacramento, CA, USA
| | - S J Emerson
- Center for Surgical Bioengineering, Deparment of Surgery, University of California Davis School of Medicine, Sacramento, CA, USA; Institute for Pediatric Regenerative Medicine, Shriners Children's Northern California, Sacramento, CA, USA
| | - S Atis
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, Université de Montpellier, Institut Régional Du Cancer de Montpellier (ICM), Montpellier, France
| | - C Benedetti
- Montpellier Ressources Imagerie, BioCampus, University of Montpellier, CNRS, INSERM, Montpellier, France
| | - S Langlois
- Montpellier Ressources Imagerie, BioCampus, University of Montpellier, CNRS, INSERM, Montpellier, France
| | - A Marquant
- ICGM, Montpellier University, CNRS, ENSCM, Montpellier, France
| | - J Prakash
- Department of Advanced Organ Bioengineering and Therapeutics, Engineered Therapeutics Section, Technical Medical Centre, University of Twente, 7500 AE, Enschede, the Netherlands
| | - A Wang
- Center for Surgical Bioengineering, Deparment of Surgery, University of California Davis School of Medicine, Sacramento, CA, USA; Institute for Pediatric Regenerative Medicine, Shriners Children's Northern California, Sacramento, CA, USA
| | - J M Devoisselle
- ICGM, Montpellier University, CNRS, ENSCM, Montpellier, France
| | - T Montier
- Univ Brest, INSERM, EFS, UMR 1078, GGB, F-29200, Brest, France; CHU de Brest, Service de Génétique Médicale et de Biologie de La Reproduction, Centre de Référence des Maladies Rares Maladies Neuromusculaires, 29200, Brest, France
| | - F Djouad
- IRMB, University of Montpellier, INSERM, 34295, Montpellier, France; Clinical Immunology and Osteoarticular Disease Therapeutic Unit, Department of Rheumatology, CHU Montpellier, 34095, Montpellier, France
| | - J P Pouget
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, Université de Montpellier, Institut Régional Du Cancer de Montpellier (ICM), Montpellier, France
| | - V Lapinte
- ICGM, Montpellier University, CNRS, ENSCM, Montpellier, France
| | - Marie Morille
- ICGM, Montpellier University, CNRS, ENSCM, Montpellier, France; Institut Universitaire de France (IUF), Paris, France.
| |
Collapse
|
4
|
Bergqvist M, Lässer C, Crescitelli R, Park K, Lötvall J. A Non-Centrifugation Method to Concentrate and Purify Extracellular Vesicles Using Superabsorbent Polymer Followed by Size Exclusion Chromatography. J Extracell Vesicles 2025; 14:e70037. [PMID: 39840900 PMCID: PMC11752139 DOI: 10.1002/jev2.70037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Accepted: 12/27/2024] [Indexed: 01/23/2025] Open
Abstract
Extracellular vesicles (EVs) can be isolated and purified from cell cultures and biofluids using different methodologies. Here, we explored a novel EV isolation approach by combining superabsorbent polymers (SAP) in a dialysis membrane with size exclusion chromatography (SEC) to achieve high concentration and purity of EVs without the use of ultracentrifugation (UC). Suspension HEK293 cells transfected with CD63 coupled with Thermo Luciferase were used to quantify the EV yield and purity. The 500 mL conditioned medium volume was initially reduced by pressure ultrafiltration, followed by UC, SAP or a centrifugal filter unit (CFU). Using either of these methods, the EVs were concentrated to a final volume of approximately 1 mL, with retained functionality. The yield, quantified by luciferase activity, was highest with UC (70%-80%), followed by SAP (60%-70%) and CFU (50%-60%). Further purification of the EVs was performed by iodixanol density cushion (IDC) or SEC (Sepharose CL-2B or 6B, in either 10 or 20 mL columns). Although the IDC and Sepharose CL-2B (10 mL) achieved the highest yields, the purity was slightly higher (30%) with IDC. In conclusion, combining SAP concentration with CL-2B SEC is an alternative and efficient way to isolate EVs without using UC.
Collapse
Affiliation(s)
- Markus Bergqvist
- Department of Internal Medicine and Clinical Nutrition, Krefting Research Centre, Institute of Medicine, Sahlgrenska Academy, University of GothenburgGothenburgSweden
| | - Cecilia Lässer
- Department of Internal Medicine and Clinical Nutrition, Krefting Research Centre, Institute of Medicine, Sahlgrenska Academy, University of GothenburgGothenburgSweden
| | - Rossella Crescitelli
- Department of Surgery, Sahlgrenska Center for Cancer Research and Wallenberg Centre for Molecular and Translational Medicine, Institute of Clinical Sciences, Sahlgrenska Academy, University of GothenburgGothenburgSweden
| | - Kyong‐Su Park
- Department of Internal Medicine and Clinical Nutrition, Krefting Research Centre, Institute of Medicine, Sahlgrenska Academy, University of GothenburgGothenburgSweden
| | - Jan Lötvall
- Department of Internal Medicine and Clinical Nutrition, Krefting Research Centre, Institute of Medicine, Sahlgrenska Academy, University of GothenburgGothenburgSweden
| |
Collapse
|
5
|
Zeng W, Zheng L, Li Y, Yang J, Mao T, Zhang J, Liu Y, Ning J, Zhang T, Huang H, Chen X, Lu F. Engineered extracellular vesicles for delivering functional Cas9/gRNA to eliminate hepatitis B virus cccDNA and integration. Emerg Microbes Infect 2024; 13:2284286. [PMID: 37982370 PMCID: PMC10763861 DOI: 10.1080/22221751.2023.2284286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 11/13/2023] [Indexed: 11/21/2023]
Abstract
The persistence of HBV covalently closed circular DNA (cccDNA) and HBV integration into the host genome in infected hepatocytes pose significant challenges to the cure of chronic HBV infection. Although CRISPR/Cas9-mediated genome editing shows promise for targeted clearance of viral genomes, a safe and efficient delivery method is currently lacking. Here, we developed a novel approach by combining light-induced heterodimerization and protein acylation to enhance the loading efficiency of Cas9 protein into extracellular vesicles (EVs). Moreover, vesicular stomatitis virus-glycoprotein (VSV-G) was incorporated onto the EVs membrane, significantly facilitating the endosomal escape of Cas9 protein and increasing its gene editing activity in recipient cells. Our results demonstrated that engineered EVs containing Cas9/gRNA and VSV-G can effectively reduce viral antigens and cccDNA levels in the HBV-replicating and infected cell models. Notably, we also confirmed the antiviral activity and high safety of the engineered EVs in the HBV-replicating mouse model generated by hydrodynamic injection and the HBV transgenic mouse model. In conclusion, engineered EVs could successfully mediate functional CRISPR/Cas9 delivery both in vitro and in vivo, leading to the clearance of episomal cccDNA and integrated viral DNA fragments, and providing a novel therapeutic approach for curing chronic HBV infection.
Collapse
Affiliation(s)
- Wanjia Zeng
- Department of Microbiology and Infectious Disease Center, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, People’s Republic of China
| | - Liwei Zheng
- Department of Microbiology and Infectious Disease Center, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, People’s Republic of China
| | - Yukun Li
- Department of Microbiology and Infectious Disease Center, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, People’s Republic of China
| | - Jing Yang
- School of Medicine, Shihezi University, Shihezi, People’s Republic of China
| | - Tianhao Mao
- Department of Microbiology and Infectious Disease Center, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, People’s Republic of China
| | - Jing Zhang
- Department of Microbiology and Infectious Disease Center, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, People’s Republic of China
| | - Yanna Liu
- Department of Gastroenterology and Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Jing Ning
- Department of Gastroenterology, Beijing Key Laboratory for Helicobacter Pylori Infection and Upper Gastrointestinal Diseases, Peking University Third Hospital, Beijing, People’s Republic of China
| | - Ting Zhang
- Department of Microbiology and Infectious Disease Center, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, People’s Republic of China
| | - Hongxin Huang
- Department of Pathogen Biology and Biosecurity, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, People’s Republic of China
| | - Xiangmei Chen
- Department of Microbiology and Infectious Disease Center, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, People’s Republic of China
| | - Fengmin Lu
- Department of Microbiology and Infectious Disease Center, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, People’s Republic of China
| |
Collapse
|
6
|
Li C, Enciso-Martinez A, Koning RI, Shahsavari M, Ten Dijke P. TGF-β regulates the release of breast cancer cell-derived extracellular vesicles and the sorting of their protein cargo by downregulating RAB27B expression. J Extracell Vesicles 2024; 13:e70026. [PMID: 39723610 DOI: 10.1002/jev2.70026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 10/24/2024] [Accepted: 11/28/2024] [Indexed: 12/28/2024] Open
Abstract
Extracellular vesicles (EVs) are important mediators of intercellular communication in the tumour microenvironment. The cytokine transforming growth factor-β (TGF-β) facilitates cancer progression via EVs secreted by cancer cells, which act on recipient cells in the tumour microenvironment. However, the mechanisms of how TGF-β affects cancer cell EV release and composition are incompletely understood. Here, we systematically investigate the effects of TGF-β on the release and protein composition of EVs from breast cancer cells. TGF-β suppresses the transcription of RAB27B mediated by SMAD3 and thereby hampers EV release. Using click chemistry and quantitative proteomics, we found that TGF-β increases the quantity of protein cargo and changes the composition of EVs by downregulating RAB27B expression. The recomposed EVs, induced by TGF-β or RAB27B depletion, inhibit CD8+ T cell-mediated breast cancer killing. Our findings reveal the critical roles of TGF-β and RAB27B in cancer development by regulating EV release and composition and thus provide potential targets to improve cancer immunotherapy.
Collapse
Affiliation(s)
- Chao Li
- Oncode Institute and Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, Netherlands
| | - Agustin Enciso-Martinez
- Oncode Institute and Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, Netherlands
- Biomedical Engineering & Physics; Laboratory of Experimental Clinical Chemistry, Laboratory Specialized Diagnostics & Research, Department of Laboratory Medicine, Amsterdam University Medical Center, Meibergdreef 9, Amsterdam, Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, Netherlands
| | - Roman I Koning
- Electron Microscopy Facility, Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, Netherlands
| | - Mona Shahsavari
- Biomedical Engineering & Physics; Laboratory of Experimental Clinical Chemistry, Laboratory Specialized Diagnostics & Research, Department of Laboratory Medicine, Amsterdam University Medical Center, Meibergdreef 9, Amsterdam, Netherlands
- Amsterdam Cardiovascular Sciences, Atherosclerosis and Ischemic Syndromes, Amsterdam, Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, Netherlands
| | - Peter Ten Dijke
- Oncode Institute and Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
7
|
Bader J, Brigger F, Leroux JC. Extracellular vesicles versus lipid nanoparticles for the delivery of nucleic acids. Adv Drug Deliv Rev 2024; 215:115461. [PMID: 39490384 DOI: 10.1016/j.addr.2024.115461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 10/21/2024] [Accepted: 10/23/2024] [Indexed: 11/05/2024]
Abstract
Extracellular vesicles (EVs) are increasingly investigated for delivering nucleic acid (NA) therapeutics, leveraging their natural role in transporting NA and protein-based cargo in cell-to-cell signaling. Their synthetic counterparts, lipid nanoparticles (LNPs), have been developed over the past decades as NA carriers, culminating in the approval of several marketed formulations such as patisiran/Onpattro® and the mRNA-1273/BNT162 COVID-19 vaccines. The success of LNPs has sparked efforts to develop innovative technologies to target extrahepatic organs, and to deliver novel therapeutic modalities, such as tools for in vivo gene editing. Fueled by the recent advancements in both fields, this review aims to provide a comprehensive overview of the basic characteristics of EV and LNP-based NA delivery systems, from EV biogenesis to structural properties of LNPs. It addresses the primary challenges encountered in utilizing these nanocarriers from a drug formulation and delivery perspective. Additionally, biodistribution profiles, in vitro and in vivo transfection outcomes, as well as their status in clinical trials are compared. Overall, this review provides insights into promising research avenues and potential dead ends for EV and LNP-based NA delivery systems.
Collapse
Affiliation(s)
- Johannes Bader
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, 8093 Zurich, Switzerland
| | - Finn Brigger
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, 8093 Zurich, Switzerland
| | - Jean-Christophe Leroux
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, 8093 Zurich, Switzerland.
| |
Collapse
|
8
|
Visser C, Rivieccio F, Krüger T, Schmidt F, Cseresnyés Z, Rohde M, Figge MT, Kniemeyer O, Blango MG, Brakhage AA. Tracking the uptake of labelled host-derived extracellular vesicles by the human fungal pathogen Aspergillus fumigatus. MICROLIFE 2024; 5:uqae022. [PMID: 39660046 PMCID: PMC11631206 DOI: 10.1093/femsml/uqae022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 10/16/2024] [Accepted: 11/04/2024] [Indexed: 12/12/2024]
Abstract
Extracellular vesicles (EVs) have gained attention as facilitators of intercellular as well as interkingdom communication during host-microbe interactions. Recently we showed that upon infection, host polymorphonuclear leukocytes produce antifungal EVs targeting the clinically important fungal pathogen Aspergillus fumigatus; however, the small size of EVs (<1 µm) complicates their functional analysis. Here, we employed a more tractable, reporter-based system to label host alveolar epithelial cell-derived EVs and enable their visualization during in vitro A. fumigatus interaction. Fusion of EV marker proteins (CD63, CD9, and CD81) with a Nanoluciferase (NLuc) and a green fluorescent protein (GFP) facilitated their relative quantification by luminescence and visualization by a fluorescence signal. The use of an NLuc fused with a GFP is advantageous as it allows for quantification and visualization of EVs simultaneously without additional external manipulation and to distinguish subpopulations of EVs. Using this system, visualization and tracking of EVs was possible using confocal laser scanning microscopy and advanced imaging analysis. These experiments revealed the propensity of host cell-derived EVs to associate with the fungal cell wall and ultimately colocalize with the cell membrane of A. fumigatus hyphae in large numbers. In conclusion, we have created a series of tools to better define the complex interplay of host-derived EVs with microbial pathogens.
Collapse
Affiliation(s)
- Corissa Visser
- Department of Molecular and Applied Microbiology, Leibniz Institute for Natural Product Research and Infection Biology—Hans Knöll Institute (Leibniz-HKI), 07745 Jena, Germany
- Institute of Microbiology, Friedrich Schiller University, 07743 Jena, Germany
| | - Flora Rivieccio
- Department of Molecular and Applied Microbiology, Leibniz Institute for Natural Product Research and Infection Biology—Hans Knöll Institute (Leibniz-HKI), 07745 Jena, Germany
- Institute of Microbiology, Friedrich Schiller University, 07743 Jena, Germany
| | - Thomas Krüger
- Department of Molecular and Applied Microbiology, Leibniz Institute for Natural Product Research and Infection Biology—Hans Knöll Institute (Leibniz-HKI), 07745 Jena, Germany
| | - Franziska Schmidt
- Department of Molecular and Applied Microbiology, Leibniz Institute for Natural Product Research and Infection Biology—Hans Knöll Institute (Leibniz-HKI), 07745 Jena, Germany
- Institute of Microbiology, Friedrich Schiller University, 07743 Jena, Germany
| | - Zoltán Cseresnyés
- Research Group Applied Systems Biology, Leibniz Institute for Natural Product Research and Infection Biology (HKI), 07745 Jena, Germany
| | - Manfred Rohde
- Helmholtz Centre for Infection Research (HZI), 38124 Braunschweig, Germany
| | - Marc Thilo Figge
- Institute of Microbiology, Friedrich Schiller University, 07743 Jena, Germany
- Research Group Applied Systems Biology, Leibniz Institute for Natural Product Research and Infection Biology (HKI), 07745 Jena, Germany
- Excellence Cluster Balance of the Microverse, Friedrich Schiller University, 07743 Jena, Germany
| | - Olaf Kniemeyer
- Department of Molecular and Applied Microbiology, Leibniz Institute for Natural Product Research and Infection Biology—Hans Knöll Institute (Leibniz-HKI), 07745 Jena, Germany
| | - Matthew G Blango
- Junior Research Group RNA Biology of Fungal Infections, Leibniz Institute for Natural Product Research and Infection Biology (Leibniz-HKI), 07745 Jena, Germany
| | - Axel A Brakhage
- Department of Molecular and Applied Microbiology, Leibniz Institute for Natural Product Research and Infection Biology—Hans Knöll Institute (Leibniz-HKI), 07745 Jena, Germany
- Institute of Microbiology, Friedrich Schiller University, 07743 Jena, Germany
- Excellence Cluster Balance of the Microverse, Friedrich Schiller University, 07743 Jena, Germany
| |
Collapse
|
9
|
Zickler AM, Liang X, Gupta D, Mamand DR, De Luca M, Corso G, Errichelli L, Hean J, Sen T, Elsharkasy OM, Kamei N, Niu Z, Zhou G, Zhou H, Roudi S, Wiklander OPB, Görgens A, Nordin JZ, Castilla‐Llorente V, EL Andaloussi S. Novel Endogenous Engineering Platform for Robust Loading and Delivery of Functional mRNA by Extracellular Vesicles. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2407619. [PMID: 39246205 PMCID: PMC11558116 DOI: 10.1002/advs.202407619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Revised: 08/27/2024] [Indexed: 09/10/2024]
Abstract
Messenger RNA (mRNA) has emerged as an attractive therapeutic molecule for a plethora of clinical applications. For in vivo functionality, mRNA therapeutics require encapsulation into effective, stable, and safe delivery systems to protect the cargo from degradation and reduce immunogenicity. Here, a bioengineering platform for efficient mRNA loading and functional delivery using bionormal nanoparticles, extracellular vesicles (EVs), is established by expressing a highly specific RNA-binding domain fused to CD63 in EV producer cells stably expressing the target mRNA. The additional combination with a fusogenic endosomal escape moiety, Vesicular Stomatitis Virus Glycoprotein, enables functional mRNA delivery in vivo at doses substantially lower than currently used clinically with synthetic lipid-based nanoparticles. Importantly, the application of EVs loaded with effective cancer immunotherapy proves highly effective in an aggressive melanoma mouse model. This technology addresses substantial drawbacks currently associated with EV-based nucleic acid delivery systems and is a leap forward to clinical EV applications.
Collapse
Affiliation(s)
- Antje M. Zickler
- Division of Biomolecular and Cellular MedicineDepartment of Laboratory MedicineKarolinska InstitutetANA FuturaAlfred‐Nobels‐Allé 8, HuddingeStockholm14152Sweden
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST)Karolinska University HospitalStockholm14186Sweden
- Karolinska ATMP Center, Karolinska InstitutetANA FuturaAlfred‐Nobels‐Allé 8, HuddingeStockholm14152Sweden
| | - Xiuming Liang
- Division of Biomolecular and Cellular MedicineDepartment of Laboratory MedicineKarolinska InstitutetANA FuturaAlfred‐Nobels‐Allé 8, HuddingeStockholm14152Sweden
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST)Karolinska University HospitalStockholm14186Sweden
- Karolinska ATMP Center, Karolinska InstitutetANA FuturaAlfred‐Nobels‐Allé 8, HuddingeStockholm14152Sweden
- Cancer Research LaboratoryShandong University‐Karolinska Institutet collaborative LaboratorySchool of Basic Medical ScienceShandong UniversityNo. 44, Wenhua Xi RoadJi'nanShandong250012P. R. China
| | - Dhanu Gupta
- Division of Biomolecular and Cellular MedicineDepartment of Laboratory MedicineKarolinska InstitutetANA FuturaAlfred‐Nobels‐Allé 8, HuddingeStockholm14152Sweden
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST)Karolinska University HospitalStockholm14186Sweden
- Institute of Developmental and Regenerative Medicine, Department of Paediatrics.University of OxfordOld Road Campus, Roosevelt Dr, HeadingtonOxfordOX3 7TYUK
| | - Doste R. Mamand
- Division of Biomolecular and Cellular MedicineDepartment of Laboratory MedicineKarolinska InstitutetANA FuturaAlfred‐Nobels‐Allé 8, HuddingeStockholm14152Sweden
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST)Karolinska University HospitalStockholm14186Sweden
- Karolinska ATMP Center, Karolinska InstitutetANA FuturaAlfred‐Nobels‐Allé 8, HuddingeStockholm14152Sweden
- Breast Center, Karolinska Comprehensive Cancer CenterKarolinska University HospitalStockholm14186Sweden
| | - Mariacristina De Luca
- Evox Therapeutics Ltd.Oxford Science ParkMedawar CentreRobert Robinson AvenueOxfordOX4 4HGUK
- Human TechnopoleViale Rita Levi Montalcini, 1Milan20157Italy
| | - Giulia Corso
- Division of Biomolecular and Cellular MedicineDepartment of Laboratory MedicineKarolinska InstitutetANA FuturaAlfred‐Nobels‐Allé 8, HuddingeStockholm14152Sweden
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST)Karolinska University HospitalStockholm14186Sweden
- Evercyte GmbHLeberstrasse 20Vienna1110Austria
| | - Lorenzo Errichelli
- Evox Therapeutics Ltd.Oxford Science ParkMedawar CentreRobert Robinson AvenueOxfordOX4 4HGUK
| | - Justin Hean
- Evox Therapeutics Ltd.Oxford Science ParkMedawar CentreRobert Robinson AvenueOxfordOX4 4HGUK
| | - Titash Sen
- Evox Therapeutics Ltd.Oxford Science ParkMedawar CentreRobert Robinson AvenueOxfordOX4 4HGUK
- Lonza BiologicsChesterford Research ParkCambridgeCB10 1XLUK
| | - Omnia M. Elsharkasy
- Division of Biomolecular and Cellular MedicineDepartment of Laboratory MedicineKarolinska InstitutetANA FuturaAlfred‐Nobels‐Allé 8, HuddingeStockholm14152Sweden
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST)Karolinska University HospitalStockholm14186Sweden
- Karolinska ATMP Center, Karolinska InstitutetANA FuturaAlfred‐Nobels‐Allé 8, HuddingeStockholm14152Sweden
| | - Noriyasu Kamei
- Division of Biomolecular and Cellular MedicineDepartment of Laboratory MedicineKarolinska InstitutetANA FuturaAlfred‐Nobels‐Allé 8, HuddingeStockholm14152Sweden
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST)Karolinska University HospitalStockholm14186Sweden
- Laboratory of Drug Delivery SystemsFaculty of Pharmaceutical SciencesKobe Gakuin University1‐1‐3 Minatojima, Chuo‐kuKobeHyogo650‐8586Japan
| | - Zheyu Niu
- Division of Biomolecular and Cellular MedicineDepartment of Laboratory MedicineKarolinska InstitutetANA FuturaAlfred‐Nobels‐Allé 8, HuddingeStockholm14152Sweden
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST)Karolinska University HospitalStockholm14186Sweden
- Department of Hepatobiliary SurgeryShandong Provincial Hospital Affiliated to Shandong First Medical UniversityNo. 324, Five Jing RoadJi'nanShandong250012P. R. China
| | - Guannan Zhou
- Division of Biomolecular and Cellular MedicineDepartment of Laboratory MedicineKarolinska InstitutetANA FuturaAlfred‐Nobels‐Allé 8, HuddingeStockholm14152Sweden
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST)Karolinska University HospitalStockholm14186Sweden
- Department of GynecologyThe Obstetrics and Gynecology Hospital of Fudan UniversityNo. 419, Fangxie RoadShanghai200011P. R. China
| | - Houze Zhou
- Division of Biomolecular and Cellular MedicineDepartment of Laboratory MedicineKarolinska InstitutetANA FuturaAlfred‐Nobels‐Allé 8, HuddingeStockholm14152Sweden
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST)Karolinska University HospitalStockholm14186Sweden
- Karolinska ATMP Center, Karolinska InstitutetANA FuturaAlfred‐Nobels‐Allé 8, HuddingeStockholm14152Sweden
| | - Samantha Roudi
- Division of Biomolecular and Cellular MedicineDepartment of Laboratory MedicineKarolinska InstitutetANA FuturaAlfred‐Nobels‐Allé 8, HuddingeStockholm14152Sweden
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST)Karolinska University HospitalStockholm14186Sweden
- Karolinska ATMP Center, Karolinska InstitutetANA FuturaAlfred‐Nobels‐Allé 8, HuddingeStockholm14152Sweden
| | - Oscar P. B. Wiklander
- Division of Biomolecular and Cellular MedicineDepartment of Laboratory MedicineKarolinska InstitutetANA FuturaAlfred‐Nobels‐Allé 8, HuddingeStockholm14152Sweden
- Karolinska ATMP Center, Karolinska InstitutetANA FuturaAlfred‐Nobels‐Allé 8, HuddingeStockholm14152Sweden
- Breast Center, Karolinska Comprehensive Cancer CenterKarolinska University HospitalStockholm14186Sweden
| | - André Görgens
- Division of Biomolecular and Cellular MedicineDepartment of Laboratory MedicineKarolinska InstitutetANA FuturaAlfred‐Nobels‐Allé 8, HuddingeStockholm14152Sweden
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST)Karolinska University HospitalStockholm14186Sweden
- Karolinska ATMP Center, Karolinska InstitutetANA FuturaAlfred‐Nobels‐Allé 8, HuddingeStockholm14152Sweden
- Institute for Transfusion MedicineUniversity Hospital EssenUniversity of Duisburg‐Essen45147EssenGermany
| | - Joel Z. Nordin
- Division of Biomolecular and Cellular MedicineDepartment of Laboratory MedicineKarolinska InstitutetANA FuturaAlfred‐Nobels‐Allé 8, HuddingeStockholm14152Sweden
- Karolinska ATMP Center, Karolinska InstitutetANA FuturaAlfred‐Nobels‐Allé 8, HuddingeStockholm14152Sweden
- Department of Clinical Immunology and Transfusion Medicine (KITM)Karolinska University HospitalStockholm14186Sweden
| | - Virginia Castilla‐Llorente
- Evox Therapeutics Ltd.Oxford Science ParkMedawar CentreRobert Robinson AvenueOxfordOX4 4HGUK
- Uncommon BioCambridge TechnoparkNewmarket RdCambridgeCB5 8PBUK
| | - Samir EL Andaloussi
- Division of Biomolecular and Cellular MedicineDepartment of Laboratory MedicineKarolinska InstitutetANA FuturaAlfred‐Nobels‐Allé 8, HuddingeStockholm14152Sweden
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST)Karolinska University HospitalStockholm14186Sweden
- Karolinska ATMP Center, Karolinska InstitutetANA FuturaAlfred‐Nobels‐Allé 8, HuddingeStockholm14152Sweden
| |
Collapse
|
10
|
Wiklander OPB, Mamand DR, Mohammad DK, Zheng W, Jawad Wiklander R, Sych T, Zickler AM, Liang X, Sharma H, Lavado A, Bost J, Roudi S, Corso G, Lennaárd AJ, Abedi-Valugerdi M, Mäger I, Alici E, Sezgin E, Nordin JZ, Gupta D, Görgens A, El Andaloussi S. Antibody-displaying extracellular vesicles for targeted cancer therapy. Nat Biomed Eng 2024; 8:1453-1468. [PMID: 38769158 PMCID: PMC11584392 DOI: 10.1038/s41551-024-01214-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 04/08/2024] [Indexed: 05/22/2024]
Abstract
Extracellular vesicles (EVs) function as natural delivery vectors and mediators of biological signals across tissues. Here, by leveraging these functionalities, we show that EVs decorated with an antibody-binding moiety specific for the fragment crystallizable (Fc) domain can be used as a modular delivery system for targeted cancer therapy. The Fc-EVs can be decorated with different types of immunoglobulin G antibody and thus be targeted to virtually any tissue of interest. Following optimization of the engineered EVs by screening Fc-binding and EV-sorting moieties, we show the targeting of EVs to cancer cells displaying the human epidermal receptor 2 or the programmed-death ligand 1, as well as lower tumour burden and extended survival of mice with subcutaneous melanoma tumours when systemically injected with EVs displaying an antibody for the programmed-death ligand 1 and loaded with the chemotherapeutic doxorubicin. EVs with Fc-binding domains may be adapted to display other Fc-fused proteins, bispecific antibodies and antibody-drug conjugates.
Collapse
Affiliation(s)
- Oscar P B Wiklander
- Department of Laboratory Medicine, Unit for Biomolecular and Cellular Medicine, Karolinska Institutet, Stockholm, Sweden.
- Breast Center, Karolinska Comprehensive Cancer Center, Karolinska University Hospital, Stockholm, Sweden.
- Karolinska ATMP Center, ANA Futura, Huddinge, Sweden.
| | - Doste R Mamand
- Department of Laboratory Medicine, Unit for Biomolecular and Cellular Medicine, Karolinska Institutet, Stockholm, Sweden
- Breast Center, Karolinska Comprehensive Cancer Center, Karolinska University Hospital, Stockholm, Sweden
- Karolinska ATMP Center, ANA Futura, Huddinge, Sweden
| | - Dara K Mohammad
- Department of Medicine Huddinge, Karolinska Institute, Stockholm, Sweden
- College of Agricultural Engineering Sciences, Salahaddin University-Erbil, Erbil, Iraq
| | - Wenyi Zheng
- Department of Laboratory Medicine, Unit for Biomolecular and Cellular Medicine, Karolinska Institutet, Stockholm, Sweden
- Karolinska ATMP Center, ANA Futura, Huddinge, Sweden
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST), Karolinska University Hospital, Huddinge, Sweden
| | - Rim Jawad Wiklander
- Department of Laboratory Medicine, Unit for Biomolecular and Cellular Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST), Karolinska University Hospital, Huddinge, Sweden
| | - Taras Sych
- Science for Life Laboratory, Department of Women's and Children's Health, Karolinska Institutet, Solna, Sweden
| | - Antje M Zickler
- Department of Laboratory Medicine, Unit for Biomolecular and Cellular Medicine, Karolinska Institutet, Stockholm, Sweden
- Karolinska ATMP Center, ANA Futura, Huddinge, Sweden
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST), Karolinska University Hospital, Huddinge, Sweden
| | - Xiuming Liang
- Department of Laboratory Medicine, Unit for Biomolecular and Cellular Medicine, Karolinska Institutet, Stockholm, Sweden
- Karolinska ATMP Center, ANA Futura, Huddinge, Sweden
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST), Karolinska University Hospital, Huddinge, Sweden
| | | | | | - Jeremy Bost
- Department of Laboratory Medicine, Unit for Biomolecular and Cellular Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST), Karolinska University Hospital, Huddinge, Sweden
| | - Samantha Roudi
- Department of Laboratory Medicine, Unit for Biomolecular and Cellular Medicine, Karolinska Institutet, Stockholm, Sweden
- Karolinska ATMP Center, ANA Futura, Huddinge, Sweden
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST), Karolinska University Hospital, Huddinge, Sweden
| | - Giulia Corso
- Department of Laboratory Medicine, Unit for Biomolecular and Cellular Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST), Karolinska University Hospital, Huddinge, Sweden
| | - Angus J Lennaárd
- Department of Laboratory Medicine, Unit for Biomolecular and Cellular Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST), Karolinska University Hospital, Huddinge, Sweden
| | - Manuchehr Abedi-Valugerdi
- Department of Laboratory Medicine, Unit for Biomolecular and Cellular Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST), Karolinska University Hospital, Huddinge, Sweden
| | - Imre Mäger
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Evren Alici
- Department of Medicine Huddinge, Karolinska Institute, Stockholm, Sweden
- Hematology Center, Karolinska University Hospital, Stockholm, Sweden
| | - Erdinc Sezgin
- Science for Life Laboratory, Department of Women's and Children's Health, Karolinska Institutet, Solna, Sweden
| | - Joel Z Nordin
- Department of Laboratory Medicine, Unit for Biomolecular and Cellular Medicine, Karolinska Institutet, Stockholm, Sweden
- Karolinska ATMP Center, ANA Futura, Huddinge, Sweden
- Department of Clinical Immunology and Transfusion Medicine (KITM), Karolinska University Hospital, Stockholm, Sweden
| | - Dhanu Gupta
- Department of Laboratory Medicine, Unit for Biomolecular and Cellular Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST), Karolinska University Hospital, Huddinge, Sweden
- Department of Paediatrics, University of Oxford, Oxford, UK
| | - André Görgens
- Department of Laboratory Medicine, Unit for Biomolecular and Cellular Medicine, Karolinska Institutet, Stockholm, Sweden
- Karolinska ATMP Center, ANA Futura, Huddinge, Sweden
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST), Karolinska University Hospital, Huddinge, Sweden
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, Duisburg, Germany
| | - Samir El Andaloussi
- Department of Laboratory Medicine, Unit for Biomolecular and Cellular Medicine, Karolinska Institutet, Stockholm, Sweden.
- Karolinska ATMP Center, ANA Futura, Huddinge, Sweden.
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST), Karolinska University Hospital, Huddinge, Sweden.
| |
Collapse
|
11
|
Viola M, Bebelman MP, Maas RGC, de Voogt WS, Verweij FJ, Seinen CS, de Jager SCA, Vader P, Pegtel DM, Petrus Gerardus Sluijter J. Hypoxia and TNF-alpha modulate extracellular vesicle release from human induced pluripotent stem cell-derived cardiomyocytes. J Extracell Vesicles 2024; 13:e70000. [PMID: 39508403 PMCID: PMC11541862 DOI: 10.1002/jev2.70000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 08/28/2024] [Accepted: 10/06/2024] [Indexed: 11/15/2024] Open
Abstract
Extracellular vesicles (EVs) have emerged as important mediators of intercellular communication in the heart under homeostatic and pathological conditions, such as myocardial infarction (MI). However, the basic mechanisms driving cardiomyocyte-derived EV (CM-EV) production following stress are poorly understood. In this study, we generated human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) that express NanoLuc-tetraspanin reporters. These modified hiPSC-CMs allow for quantification of tetraspanin-positive CM-EV secretion from small numbers of cells without the need for time-consuming EV isolation techniques. We subjected these cells to a panel of small molecules to study their effect on CM-EV biogenesis and secretion under basal and stress-associated conditions. We observed that EV biogenesis is context-dependent in hiPSC-CMs. Nutrient starvation decreases CM-EV secretion while hypoxia increases the production of CM-EVs in a nSmase2-dependent manner. Moreover, the inflammatory cytokine TNF-α increased CM-EV secretion through a process involving NLRP3 inflammasome activation and mTOR signalling. Here, we detailed for the first time the regulatory mechanisms of EV biogenesis in hiPSC-CMs upon MI-associated stressors.
Collapse
Affiliation(s)
- Margarida Viola
- Department of Experimental Cardiology, University Medical Center UtrechtUtrecht UniversityUtrechtThe Netherlands
| | - Maarten P. Bebelman
- Department of Pathology, Cancer Center AmsterdamVU University Medical CenterAmsterdamThe Netherlands
| | - Renee G. C. Maas
- Department of Experimental Cardiology, University Medical Center UtrechtUtrecht UniversityUtrechtThe Netherlands
| | | | - Frederik J. Verweij
- Division of Cell Biology, Neurobiology and BiophysicsUtrecht UniversityUtrechtThe Netherlands
| | - Cor S. Seinen
- CDL ResearchUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Saskia C. A. de Jager
- Department of Experimental Cardiology, University Medical Center UtrechtUtrecht UniversityUtrechtThe Netherlands
| | - Pieter Vader
- Department of Experimental Cardiology, University Medical Center UtrechtUtrecht UniversityUtrechtThe Netherlands
- CDL ResearchUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Dirk Michiel Pegtel
- Department of Pathology, Cancer Center AmsterdamVU University Medical CenterAmsterdamThe Netherlands
| | | |
Collapse
|
12
|
Bobbili MR, Görgens A, Yan Y, Vogt S, Gupta D, Corso G, Barbaria S, Patrioli C, Weilner S, Pultar M, Jacak J, Hackl M, Schosserer M, Grillari R, Kjems J, Andaloussi SEL, Grillari J. Snorkel-tag based affinity chromatography for recombinant extracellular vesicle purification. J Extracell Vesicles 2024; 13:e12523. [PMID: 39400515 PMCID: PMC11472238 DOI: 10.1002/jev2.12523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 09/12/2024] [Indexed: 10/15/2024] Open
Abstract
Extracellular vesicles (EVs) are lipid nanoparticles and play an important role in cell-cell communications, making them potential therapeutic agents and allowing to engineer for targeted drug delivery. The expanding applications of EVs in next generation medicine is still limited by existing tools for scaling standardized EV production, single EV tracing and analytics, and thus provide only a snapshot of tissue-specific EV cargo information. Here, we present the Snorkel-tag, for which we have genetically fused the EV surface marker protein CD81, to a series of tags with an additional transmembrane domain to be displayed on the EV surface, resembling a snorkel. This system enables the affinity purification of EVs from complex matrices in a non-destructive form while maintaining EV characteristics in terms of surface protein profiles, associated miRNA patterns and uptake into a model cell line. Therefore, we consider the Snorkel-tag to be a widely applicable tool in EV research, allowing for efficient preparation of EV standards and reference materials, or dissecting EVs with different surface markers when fusing to other tetraspanins in vitro or in vivo.
Collapse
Affiliation(s)
- Madhusudhan Reddy Bobbili
- Institute of Molecular Biotechnology, Department of BiotechnologyBOKU UniversityViennaAustria
- Ludwig Boltzmann Institute for TraumatologyThe Research Center in Cooperation with AUVAViennaAustria
- Austrian Cluster for Tissue Regeneration
| | - André Görgens
- Department of Laboratory Medicine, Division of Biomolecular and Cellular MedicineKarolinska InstitutetStockholmSweden
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST)Karolinska University Hospital Huddinge and Karolinska Comprehensive Cancer CenterStockholmSweden
- Institute for Transfusion Medicine, University Hospital EssenUniversity of Duisburg‐EssenEssenGermany
| | - Yan Yan
- Department of Molecular Biology and Genetics, Centre for Cellular Signal Patterns (CellPat), Interdisciplinary Nanoscience Centre (iNANO)Aarhus UniversityAarhus CDenmark
- Omiics ApSAarhus NDenmark
| | - Stefan Vogt
- Institute of Molecular Biotechnology, Department of BiotechnologyBOKU UniversityViennaAustria
| | - Dhanu Gupta
- Department of Laboratory Medicine, Division of Biomolecular and Cellular MedicineKarolinska InstitutetStockholmSweden
- Institute of Developmental and Regenerative MedicineUniversity of Oxford, IMS‐Tetsuya Nakamura Building, Old Road Campus, Roosevelt Dr, HeadingtonOxfordUnited Kingdom
- Department of PaediatricsUniversity of Oxford, South Parks RoadOxfordUnited Kingdom
| | - Giulia Corso
- Department of Laboratory Medicine, Division of Biomolecular and Cellular MedicineKarolinska InstitutetStockholmSweden
- Evercyte GmbHViennaAustria
| | - Samir Barbaria
- Institute of Molecular Biotechnology, Department of BiotechnologyBOKU UniversityViennaAustria
| | - Carolina Patrioli
- Institute of Molecular Biotechnology, Department of BiotechnologyBOKU UniversityViennaAustria
| | - Sylvia Weilner
- Institute of Molecular Biotechnology, Department of BiotechnologyBOKU UniversityViennaAustria
| | | | - Jaroslaw Jacak
- Ludwig Boltzmann Institute for TraumatologyThe Research Center in Cooperation with AUVAViennaAustria
- School of Medical Engineering and Applied Social ScienceUniversity of Applied Sciences Upper AustriaLinzAustria
| | - Matthias Hackl
- Austrian Cluster for Tissue Regeneration
- TAmiRNATAmiRNA GmbHViennaAustria
| | - Markus Schosserer
- Institute of Molecular Biotechnology, Department of BiotechnologyBOKU UniversityViennaAustria
- Austrian Cluster for Tissue Regeneration
- Institute of Medical GeneticsCenter for Pathobiochemistry and GeneticsMedical University of ViennaViennaAustria
| | - Regina Grillari
- Austrian Cluster for Tissue Regeneration
- Evercyte GmbHViennaAustria
| | - Jørgen Kjems
- Department of Molecular Biology and Genetics, Centre for Cellular Signal Patterns (CellPat), Interdisciplinary Nanoscience Centre (iNANO)Aarhus UniversityAarhus CDenmark
| | - Samir EL Andaloussi
- Department of Laboratory Medicine, Division of Biomolecular and Cellular MedicineKarolinska InstitutetStockholmSweden
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST)Karolinska University Hospital Huddinge and Karolinska Comprehensive Cancer CenterStockholmSweden
| | - Johannes Grillari
- Institute of Molecular Biotechnology, Department of BiotechnologyBOKU UniversityViennaAustria
- Ludwig Boltzmann Institute for TraumatologyThe Research Center in Cooperation with AUVAViennaAustria
- Austrian Cluster for Tissue Regeneration
| |
Collapse
|
13
|
Seo GM, Lee H, Kang YJ, Kim D, Sung JH. Development of in vitro model of exosome transport in microfluidic gut-brain axis-on-a-chip. LAB ON A CHIP 2024; 24:4581-4593. [PMID: 39230477 DOI: 10.1039/d4lc00490f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
The gut communicates with the brain in a variety of ways known as the gut-brain axis (GBA), which is known to affect neurophysiological functions as well as neuronal disorders. Exosomes capable of passing through the blood-brain-barrier (BBB) have received attention as a mediator of gut-brain signaling and drug delivery vehicles. In conventional well plate-based experiments, it is difficult to observe the exosome movement in real time. Here, we developed a microfluidic-based GBA chip for co-culturing gut epithelial cells and neuronal cells and simultaneously observing exosome transport. The GBA-chip is aimed to mimic the in vivo situation of convective flow in blood vessels and convective and diffusive transport in the tissue interstitium. Here, fluorescence-labeled exosome was produced by transfection of HEK-293T cells with CD63-GFP plasmid. We observed in real time the secretion of CD63-GFP-exosomes by the transfected HEK-293T cells in the chip, and transport of the exosomes to neuronal cells and analyzed the dynamics of GFP-exosome movement. Our model is expected to enhance understanding of the roles of exosome in GBA.
Collapse
Affiliation(s)
- Gwang Myeong Seo
- Department of Chemical Engineering, Hongik University, Seoul, 04066, Republic of Korea.
| | - Hongki Lee
- School of Electrical and Electronic Engineering, Yonsei University, Seoul, Korea 03722
| | - Yeon Jae Kang
- Department of Chemical Engineering, Hongik University, Seoul, 04066, Republic of Korea.
| | - Donghyun Kim
- School of Electrical and Electronic Engineering, Yonsei University, Seoul, Korea 03722
| | - Jong Hwan Sung
- Department of Chemical Engineering, Hongik University, Seoul, 04066, Republic of Korea.
| |
Collapse
|
14
|
Chen C, Pan X, Sun M, Wang J, Su X, Zhang T, Chen Y, Wu D, Li J, Wu S, Yan X. Phospholipid-Anchored Ligand Conjugation on Extracellular Vesicles for Enhanced Cancer Targeting. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2310712. [PMID: 38733222 DOI: 10.1002/smll.202310712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 03/27/2024] [Indexed: 05/13/2024]
Abstract
Extracellular vesicles (EVs) are recognized as potential candidates for next-generation drug delivery systems. However, the inherent cancer-targeting efficiency is unsatisfactory, necessitating surface modification to attach cell-binding ligands. By utilizing phospholipase D from Streptomyces in combination with maleimide-containing primary alcohol, the authors successfully anchored ligands onto milk-derived EVs (mEVs), overcoming the issues of ligand leakage or functional alteration seen in traditional methods. Quantitative nano-flow cytometry demonstrated that over 90% of mEVs are effectively modified with hundreds to thousands of ligands. The resulting mEV formulations exhibited remarkable long-term stability in conjugation proportion, ligand number, size distribution, and particle concentration, even after months of storage. It is further shown that conjugating transferrin onto mEVs significantly enhanced cellular uptake and induced pronounced cytotoxic effects when loaded with paclitaxel. Overall, this study presents a highly efficient, stable, cost-effective, and scalable ligand conjugation approach, offering a promising strategy for targeted drug delivery of EVs.
Collapse
Affiliation(s)
- Chaoxiang Chen
- Department of Biological Engineering, College of Ocean Food and Biological Engineering, Jimei University, Xiamen, Fujian, 361021, China
| | - Xueping Pan
- Department of Biological Engineering, College of Ocean Food and Biological Engineering, Jimei University, Xiamen, Fujian, 361021, China
| | - Mengdi Sun
- Department of Biological Engineering, College of Ocean Food and Biological Engineering, Jimei University, Xiamen, Fujian, 361021, China
| | - Jialin Wang
- Department of Biological Engineering, College of Ocean Food and Biological Engineering, Jimei University, Xiamen, Fujian, 361021, China
| | - Xueqi Su
- Department of Biological Engineering, College of Ocean Food and Biological Engineering, Jimei University, Xiamen, Fujian, 361021, China
| | - Tianyu Zhang
- Department of Biological Engineering, College of Ocean Food and Biological Engineering, Jimei University, Xiamen, Fujian, 361021, China
| | - Yulei Chen
- Department of Biological Engineering, College of Ocean Food and Biological Engineering, Jimei University, Xiamen, Fujian, 361021, China
| | - Daren Wu
- Department of Biological Engineering, College of Ocean Food and Biological Engineering, Jimei University, Xiamen, Fujian, 361021, China
| | - Jian Li
- Department of Biological Engineering, College of Ocean Food and Biological Engineering, Jimei University, Xiamen, Fujian, 361021, China
| | - Shuqi Wu
- School of Life Sciences, Key Laboratory of Space Bioscience & Biotechnology, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Xiaomei Yan
- Department of Chemical Biology, MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, Key Laboratory for Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, Fujian, 361005, China
| |
Collapse
|
15
|
Gangadaran P, Khan F, Rajendran RL, Onkar A, Goenka A, Ahn B. Unveiling Invisible Extracellular Vesicles: Cutting-Edge Technologies for Their in Vivo Visualization. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2024; 16:e2009. [PMID: 39439198 PMCID: PMC11670046 DOI: 10.1002/wnan.2009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 09/11/2024] [Accepted: 10/04/2024] [Indexed: 10/25/2024]
Abstract
Extracellular vesicles (EVs), nanosized lipid bilayer vesicles released by nearly all types of cells, play pivotal roles as intercellular signaling mediators with diverse biological activities. Their adaptability has attracted interest in exploring their role as disease biomarker theranostics. However, the in vivo biodistribution and pharmacokinetic profiles of EVs, particularly following administration into living subjects, remain unclear. Thus, in vivo imaging is vital to enhance our understanding of the homing and retention patterns, blood and tissue half-life, and excretion pathways of exogenous EVs, thereby advancing real-time monitoring within biological systems and their therapeutic applications. This review examines state-of-the-art methods including EV labeling with various agents, including optical imaging, magnetic resonance imaging, and nuclear imaging. The strengths and weaknesses of each technique are comprehensively explored, emphasizing their clinical translation. Despite the potential of EVs as cancer theranostics, achieving a thorough understanding of their in vivo behavior is challenging. This review highlights the urgency of addressing current questions in the biology and therapeutic applications of EVs. It underscores the need for continued research to unravel the complexities surrounding EVs and their potential clinical implications. By identifying these challenges, this review contributes to ongoing efforts to optimize EV imaging techniques for clinical use. Ultimately, bridging the gap between research advancements and clinical applications will facilitate the integration of EV-based theranostics, marking a crucial step toward harnessing the full potential of EVs in medical practice.
Collapse
Affiliation(s)
- Prakash Gangadaran
- BK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Department of Biomedical ScienceSchool of Medicine, Kyungpook National UniversityDaeguRepublic of Korea
- Department of Nuclear MedicineSchool of Medicine, Kyungpook National UniversityDaeguRepublic of Korea
- Cardiovascular Research Institute, Kyungpook National UniversityDaeguRepublic of Korea
| | - Fatima Khan
- Department of Cancer BiologyLerner Research Institute, Cleveland ClinicClevelandOhioUSA
| | - Ramya Lakshmi Rajendran
- Department of Nuclear MedicineSchool of Medicine, Kyungpook National UniversityDaeguRepublic of Korea
- Cardiovascular Research Institute, Kyungpook National UniversityDaeguRepublic of Korea
| | - Akanksha Onkar
- Department of Laboratory MedicineUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Anshika Goenka
- Department of Hematology and Medical OncologyWinship Cancer Institute, Emory UniversityAtlantaGeorgiaUSA
| | - Byeong‐Cheol Ahn
- BK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Department of Biomedical ScienceSchool of Medicine, Kyungpook National UniversityDaeguRepublic of Korea
- Department of Nuclear MedicineSchool of Medicine, Kyungpook National UniversityDaeguRepublic of Korea
- Cardiovascular Research Institute, Kyungpook National UniversityDaeguRepublic of Korea
- Department of Nuclear MedicineKyungpook National University HospitalDaeguRepublic of Korea
| |
Collapse
|
16
|
Larsen JH, Jensen IS, Svenningsen P. Benchmarking transcriptome deconvolution methods for estimating tissue- and cell-type-specific extracellular vesicle abundances. J Extracell Vesicles 2024; 13:e12511. [PMID: 39320021 PMCID: PMC11423344 DOI: 10.1002/jev2.12511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 08/28/2024] [Indexed: 09/26/2024] Open
Abstract
Extracellular vesicles (EVs) contain cell-derived lipids, proteins and RNAs; however, determining the tissue- and cell-type-specific EV abundances in body fluids remains a significant hurdle for our understanding of EV biology. While tissue- and cell-type-specific EV abundances can be estimated by matching the EV's transcriptome to a tissue's/cell type's expression signature using deconvolutional methods, a comparative assessment of deconvolution methods' performance on EV transcriptome data is currently lacking. We benchmarked 11 deconvolution methods using data from four cell lines and their EVs, in silico mixtures, 118 human plasma and 88 urine EVs. We identified deconvolution methods that estimated cell type-specific abundances of pure and in silico mixed cell line-derived EV samples with high accuracy. Using data from two urine EV cohorts with different EV isolation procedures, four deconvolution methods produced highly similar results. The three methods were also concordant in their tissue- and cell-type-specific plasma EV abundance estimates. We identified driving factors for deconvolution accuracy and highlighted the importance of implementing biological knowledge in creating the tissue/cell type signature. Overall, our analyses demonstrate that the deconvolution algorithms DWLS and CIBERSORTx produce highly similar and accurate estimates of tissue- and cell-type-specific EV abundances in biological fluids.
Collapse
Affiliation(s)
| | - Iben Skov Jensen
- Department of Molecular MedicineUniversity of Southern DenmarkOdenseDenmark
| | - Per Svenningsen
- Department of Molecular MedicineUniversity of Southern DenmarkOdenseDenmark
| |
Collapse
|
17
|
Park DJ, Choi W, Zhang H, Eliceiri BP. Lineage Mapping of Extracellular Vesicles: What Cells Do They Come from And Where Do They Go? Adv Wound Care (New Rochelle) 2024. [PMID: 39099339 DOI: 10.1089/wound.2024.0068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/06/2024] Open
Abstract
Significance: Release of extracellular vesicles (EVs) by various cell types has been shown to mediate the delivery of biologically active payloads from donor cells to recipient cells; however, it remains unclear what cell types these EVs come from. With a focus on fluorescent reporters to monitor the release of EVs, especially those under the control of cell type-specific promoters, we address the translational relevance of genetic tools in cultured cells, normal tissues, and in models of development, injury, cancer, and wound healing. Recent Advances: It is well established that EVs are released by many cell types in the body via fusion and release processes at the plasma membrane. Since there remains debate about what fraction of EVs are released through regulated endosomal trafficking pathways versus nonspecific mechanisms, the development and validation of novel molecular tools are important to address the cellular source of EVs. Critical Issues: There is a need to develop and characterize new cell type-specific reporter mouse models that build upon the examples detailed here to identify the cellular source of EVs with genetic approaches being useful in addressing these critical limitations. Future Directions: Advances in reporter systems will drive a better understanding of EV subsets to identify compartment-specific EV localization to guide the development of more translationally relevant models for the wound healing field.
Collapse
Affiliation(s)
- Dong Jun Park
- Department of Surgery, University of California San Diego, La Jolla, California, USA
| | - Wooil Choi
- Department of Surgery, University of California San Diego, La Jolla, California, USA
| | - Hanan Zhang
- Department of Surgery, University of California San Diego, La Jolla, California, USA
| | - Brian P Eliceiri
- Department of Surgery, University of California San Diego, La Jolla, California, USA
- Department of Dermatology, University of California San Diego, La Jolla, California, USA
| |
Collapse
|
18
|
Adamo G, Santonicola P, Picciotto S, Gargano P, Nicosia A, Longo V, Aloi N, Romancino DP, Paterna A, Rao E, Raccosta S, Noto R, Salamone M, Deidda I, Costa S, Di Sano C, Zampi G, Morsbach S, Landfester K, Colombo P, Wei M, Bergese P, Touzet N, Manno M, Di Schiavi E, Bongiovanni A. Extracellular vesicles from the microalga Tetraselmis chuii are biocompatible and exhibit unique bone tropism along with antioxidant and anti-inflammatory properties. Commun Biol 2024; 7:941. [PMID: 39097626 PMCID: PMC11297973 DOI: 10.1038/s42003-024-06612-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 07/22/2024] [Indexed: 08/05/2024] Open
Abstract
Extracellular vesicles (EVs) are membrane-enclosed bio-nanoparticles secreted by cells and naturally evolved to transport various bioactive molecules between cells and even organisms. These cellular objects are considered one of the most promising bio-nanovehicles for the delivery of native and exogenous molecular cargo. However, many challenges with state-of-the-art EV-based candidates as drug carriers still exist, including issues with scalability, batch-to-batch reproducibility, and cost-sustainability of the final therapeutic formulation. Microalgal extracellular vesicles, which we named nanoalgosomes, are naturally released by various microalgal species. Here, we evaluate the innate biological properties of nanoalgosomes derived from cultures of the marine microalgae Tetraselmis chuii, using an optimized manufacturing protocol. Our investigation of nanoalgosome biocompatibility in preclinical models includes toxicological analyses, using the invertebrate model organism Caenorhabditis elegans, hematological and immunological evaluations ex vivo and in mice. We evaluate nanoalgosome cellular uptake mechanisms in C. elegans at cellular and subcellular levels, and study their biodistribution in mice with accurate space-time resolution. Further examination highlights the antioxidant and anti-inflammatory bioactivities of nanoalgosomes. This holistic approach to nanoalgosome functional characterization demonstrates that they are biocompatible and innate bioactive effectors with unique bone tropism. These findings suggest that nanoalgosomes have significant potential for future therapeutic applications.
Collapse
Affiliation(s)
- Giorgia Adamo
- Cell-Tech HUB at Institute for Research and Biomedical Innovation, National Research Council of Italy (CNR), Palermo, Italy
| | - Pamela Santonicola
- Institute of Biosciences and BioResources, National Research Council (CNR), Naples, Italy
| | - Sabrina Picciotto
- Cell-Tech HUB at Institute for Research and Biomedical Innovation, National Research Council of Italy (CNR), Palermo, Italy
| | - Paola Gargano
- Cell-Tech HUB at Institute for Research and Biomedical Innovation, National Research Council of Italy (CNR), Palermo, Italy
| | - Aldo Nicosia
- Cell-Tech HUB at Institute for Research and Biomedical Innovation, National Research Council of Italy (CNR), Palermo, Italy
| | - Valeria Longo
- Institute for Research and Biomedical Innovation, National Research Council of Italy (CNR), Palermo, Italy
| | - Noemi Aloi
- Institute for Research and Biomedical Innovation, National Research Council of Italy (CNR), Palermo, Italy
| | - Daniele P Romancino
- Cell-Tech HUB at Institute for Research and Biomedical Innovation, National Research Council of Italy (CNR), Palermo, Italy
| | - Angela Paterna
- Cell-Tech HUB at Institute of Biophysics, National Research Council of Italy (CNR), Palermo, Italy
| | - Estella Rao
- Cell-Tech HUB at Institute of Biophysics, National Research Council of Italy (CNR), Palermo, Italy
| | - Samuele Raccosta
- Cell-Tech HUB at Institute of Biophysics, National Research Council of Italy (CNR), Palermo, Italy
| | - Rosina Noto
- Cell-Tech HUB at Institute of Biophysics, National Research Council of Italy (CNR), Palermo, Italy
| | - Monica Salamone
- Cell-Tech HUB at Institute for Research and Biomedical Innovation, National Research Council of Italy (CNR), Palermo, Italy
| | - Irene Deidda
- Institute for Research and Biomedical Innovation, National Research Council of Italy (CNR), Palermo, Italy
| | - Salvatore Costa
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, Palermo, Italy
| | - Caterina Di Sano
- Institute of Translational Pharmacology, National Research Council of Italy (CNR), Palermo, Italy
| | - Giuseppina Zampi
- Institute of Biosciences and BioResources, National Research Council (CNR), Naples, Italy
| | - Svenja Morsbach
- Max Planck Institute for Polymer Research (MPIP), Mainz, Germany
| | | | - Paolo Colombo
- Institute for Research and Biomedical Innovation, National Research Council of Italy (CNR), Palermo, Italy
| | - Mingxing Wei
- Cellvax SAS, Villejuif Bio Park, 1 Mail du Professeur Georges Mathé, Villejuif, France
| | - Paolo Bergese
- Cell-Tech HUB at Institute for Research and Biomedical Innovation, National Research Council of Italy (CNR), Palermo, Italy
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
- Center for Colloid and Surface Science (CSGI), Florence, Italy
| | - Nicolas Touzet
- Department of Environmental Science, School of Science, Centre for Environmental Research, Innovation and Sustainability, CERIS, Atlantic Technological University Sligo, Sligo, Ireland
| | - Mauro Manno
- Cell-Tech HUB at Institute of Biophysics, National Research Council of Italy (CNR), Palermo, Italy
| | - Elia Di Schiavi
- Institute of Biosciences and BioResources, National Research Council (CNR), Naples, Italy
| | - Antonella Bongiovanni
- Cell-Tech HUB at Institute for Research and Biomedical Innovation, National Research Council of Italy (CNR), Palermo, Italy.
| |
Collapse
|
19
|
Xia Y, Zhang J, Liu G, Wolfram J. Immunogenicity of Extracellular Vesicles. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2403199. [PMID: 38932653 DOI: 10.1002/adma.202403199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Revised: 05/30/2024] [Indexed: 06/28/2024]
Abstract
Extracellular vesicles (EVs) are promising next-generation therapeutics and drug delivery systems due to demonstrated safety and efficacy in preclinical models and early-stage clinical trials. There is an urgent need to address the immunogenicity of EVs (beyond the apparent lack of immunotoxicity) to advance clinical development. To date, few studies have assessed unintended immunological recognition of EVs. An in-depth understanding of EV-induced immunogenicity and clearance is necessary to develop effective therapeutic strategies, including approaches to mitigate immunological recognition when undesired. This article summarizes various factors involved in the potential immunogenicity of EVs and strategies to reduce immunological recognition for improved therapeutic benefit.
Collapse
Affiliation(s)
- Yutian Xia
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang An Biomedicine Laboratory, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen, 361102, China
| | - Jianzhong Zhang
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Gang Liu
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang An Biomedicine Laboratory, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen, 361102, China
| | - Joy Wolfram
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, 4072, Australia
- School of Chemical Engineering, The University of Queensland, Brisbane, QLD, 4072, Australia
| |
Collapse
|
20
|
Liu B, Nguyen PL, Yu H, Li X, Wang H, Nguyen TGB, Sahoo PK, Sur M, Reddy J, Sillman S, Kachman SD, Altartouri B, Lu G, Natarajan SK, Pattabiraman M, Yu J. Honey vesicle-like nanoparticles protect aged liver from non-alcoholic steatohepatitis. Acta Pharm Sin B 2024; 14:3661-3679. [PMID: 39220874 PMCID: PMC11365403 DOI: 10.1016/j.apsb.2024.05.002] [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: 01/28/2024] [Revised: 04/23/2024] [Accepted: 04/30/2024] [Indexed: 09/04/2024] Open
Abstract
Non-alcoholic steatohepatitis (NASH), an advanced form of non-alcoholic fatty liver disease (NAFLD), has emerged as the leading cause of liver failure and related death. Currently, no medication is specifically approved to treat NAFLD or NASH. Here we report that oral administration of honey vesicle-like nanoparticles (H-VLNs) to naturally aged mice protects the liver from NASH development. H-VLNs are dominantly taken up by Kupffer cells in the liver and suppress hepatic chronic inflammation and further development of fibrosis and nodule formation in aged mice. Besides their reported anti-inflammasome function, H-VLNs are found to inhibit the transcriptional activities of C-JUN and nuclear factor-kappa B (NF-κB). MicroRNAs miR5119 and miR5108 and phenolic compound luteolin in H-VLNs are identified in suppressing both the C-JUN and NF-κB pathways. Collectively, oral intake of H-VLNs represents a promising new user-friendly modality to prevent the development of NASH.
Collapse
Affiliation(s)
- Baolong Liu
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, USA
- Shaanxi Key Laboratory of Molecular Biology for Agriculture, College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
| | - Phuong Linh Nguyen
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, USA
- Department of Nutrition, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Han Yu
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, USA
- Department of Nutrition, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Xingzhi Li
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, USA
| | - Huiren Wang
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, USA
| | - Tram Gia Bao Nguyen
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, USA
| | - Prakash Kumar Sahoo
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, USA
| | - Meghna Sur
- School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, USA
| | - Jay Reddy
- School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, USA
| | - Sarah Sillman
- School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, USA
| | - Stephen D. Kachman
- Department of Statistics, University of Nebraska-Lincoln, Lincoln, NE 68583, USA
| | - Bara Altartouri
- Center for Biotechnology, University of Nebraska-Lincoln, Lincoln, NE 68588, USA
| | - Guoqing Lu
- Department of Biology, University of Nebraska Omaha, Omaha, NE 68182, USA
| | - Sathish Kumar Natarajan
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, USA
| | - Mahesh Pattabiraman
- Department of Chemistry, University of Nebraska-Kearney, Kearney, NE 68849, USA
| | - Jiujiu Yu
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, USA
- Department of Nutrition, Case Western Reserve University, Cleveland, OH 44106, USA
| |
Collapse
|
21
|
Kim H, Jung SO, Lee S, Lee Y. Bioluminescent Systems for Theranostic Applications. Int J Mol Sci 2024; 25:7563. [PMID: 39062805 PMCID: PMC11277111 DOI: 10.3390/ijms25147563] [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: 05/31/2024] [Revised: 07/03/2024] [Accepted: 07/08/2024] [Indexed: 07/28/2024] Open
Abstract
Bioluminescence, the light produced by biochemical reactions involving luciferases in living organisms, has been extensively investigated for various applications. It has attracted particular interest as an internal light source for theranostic applications due to its safe and efficient characteristics that overcome the limited penetration of conventional external light sources. Recent advancements in protein engineering technologies and protein delivery platforms have expanded the application of bioluminescence to a wide range of theranostic areas, including bioimaging, biosensing, photodynamic therapy, and optogenetics. This comprehensive review presents the fundamental concepts of bioluminescence and explores its recent applications across diverse fields. Moreover, it discusses future research directions based on the current status of bioluminescent systems for further expansion of their potential.
Collapse
Affiliation(s)
- Hyemin Kim
- Department of Cosmetics Engineering, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea; (S.O.J.); (S.L.); (Y.L.)
| | | | | | | |
Collapse
|
22
|
Wardhani K, Levina A, Grau GER, Lay PA. Fluorescent, phosphorescent, magnetic resonance contrast and radioactive tracer labelling of extracellular vesicles. Chem Soc Rev 2024; 53:6779-6829. [PMID: 38828885 DOI: 10.1039/d2cs00238h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2024]
Abstract
This review focusses on the significance of fluorescent, phosphorescent labelling and tracking of extracellular vesicles (EVs) for unravelling their biology, pathophysiology, and potential diagnostic and therapeutic uses. Various labeling strategies, such as lipid membrane, surface protein, luminal, nucleic acid, radionuclide, quantum dot labels, and metal complex-based stains, are evaluated for visualizing and characterizing EVs. Direct labelling with fluorescent lipophilic dyes is simple but generally lacks specificity, while surface protein labelling offers selectivity but may affect EV-cell interactions. Luminal and nucleic acid labelling strategies have their own advantages and challenges. Each labelling approach has strengths and weaknesses, which require a suitable probe and technique based on research goals, but new tetranuclear polypyridylruthenium(II) complexes as phosphorescent probes have strong phosphorescence, selective staining, and stability. Future research should prioritize the design of novel fluorescent probes and labelling platforms that can significantly enhance the efficiency, accuracy, and specificity of EV labeling, while preserving their composition and functionality. It is crucial to reduce false positive signals and explore the potential of multimodal imaging techniques to gain comprehensive insights into EVs.
Collapse
Affiliation(s)
- Kartika Wardhani
- School of Chemistry, The University of Sydney, Sydney, New South Wales, 2006, Australia.
- Biochemistry and Biotechnology (B-TEK) Group, Bioscience Division, Los Alamos National Laboratory, Los Alamos, New Mexico, 87545, USA
| | - Aviva Levina
- School of Chemistry, The University of Sydney, Sydney, New South Wales, 2006, Australia.
| | - Georges E R Grau
- Sydney Nano, The University of Sydney, Sydney, New South Wales, 2006, Australia
- Sydney Cancer Network, The University of Sydney, Sydney, New South Wales, 2006, Australia
- Marie Bashir Institute, The University of Sydney, Sydney, New South Wales, 2006, Australia
- Vascular Immunology Unit, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, 2006, Australia
| | - Peter A Lay
- School of Chemistry, The University of Sydney, Sydney, New South Wales, 2006, Australia.
- Sydney Nano, The University of Sydney, Sydney, New South Wales, 2006, Australia
- Sydney Cancer Network, The University of Sydney, Sydney, New South Wales, 2006, Australia
- Marie Bashir Institute, The University of Sydney, Sydney, New South Wales, 2006, Australia
- Sydney Analytical, The University of Sydney, Sydney, New South Wales, 2006, Australia
| |
Collapse
|
23
|
Palmulli R, Couty M, Piontek MC, Ponnaiah M, Dingli F, Verweij FJ, Charrin S, Tantucci M, Sasidharan S, Rubinstein E, Kontush A, Loew D, Lhomme M, Roos WH, Raposo G, van Niel G. CD63 sorts cholesterol into endosomes for storage and distribution via exosomes. Nat Cell Biol 2024; 26:1093-1109. [PMID: 38886558 DOI: 10.1038/s41556-024-01432-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Accepted: 05/01/2024] [Indexed: 06/20/2024]
Abstract
Extracellular vesicles such as exosomes are now recognized as key players in intercellular communication. Their role is influenced by the specific repertoires of proteins and lipids, which are enriched when they are generated as intraluminal vesicles (ILVs) in multivesicular endosomes. Here we report that a key component of small extracellular vesicles, the tetraspanin CD63, sorts cholesterol to ILVs, generating a pool that can be mobilized by the NPC1/2 complex, and exported via exosomes to recipient cells. In the absence of CD63, cholesterol is retrieved from the endosomes by actin-dependent vesicular transport, placing CD63 and cholesterol at the centre of a balance between inward and outward budding of endomembranes. These results establish CD63 as a lipid-sorting mechanism within endosomes, and show that ILVs and exosomes are alternative providers of cholesterol.
Collapse
Affiliation(s)
- Roberta Palmulli
- Institut Curie, PSL Research University, CNRS, UMR144, 26 rue d'Ulm, 75248, Paris Cedex 05, France
- Institute of Psychiatry and Neuroscience of Paris (IPNP), Université de Paris, Paris, France
| | - Mickaël Couty
- Institute of Psychiatry and Neuroscience of Paris (IPNP), Université de Paris, Paris, France
- CRCI2NA, Nantes Université, Inserm UMR1307, CNRS UMR6075, Université d'Angers, Nantes, France
| | - Melissa C Piontek
- Moleculaire Biofysica, Zernike Instituut, Rijksuniversiteit Groningen, Groningen, The Netherlands
| | - Maharajah Ponnaiah
- Foundation for Innovation in Cardiometabolism and Nutrition (IHU ICAN, ICAN OMICS and ICAN I/O), F-75013, Paris, France
| | - Florent Dingli
- CurieCoreTech Spectrométrie de Masse Protéomique, Institut Curie, PSL Research University, Paris, France
| | - Frederik J Verweij
- Institute of Psychiatry and Neuroscience of Paris (IPNP), Université de Paris, Paris, France
| | - Stéphanie Charrin
- Centre d'Immunologie et des Maladies Infectieuses (CIMI), Sorbonne Université, Inserm, Paris, France
| | - Matteo Tantucci
- Institute of Psychiatry and Neuroscience of Paris (IPNP), Université de Paris, Paris, France
| | - Sajitha Sasidharan
- Moleculaire Biofysica, Zernike Instituut, Rijksuniversiteit Groningen, Groningen, The Netherlands
| | - Eric Rubinstein
- Centre d'Immunologie et des Maladies Infectieuses (CIMI), Sorbonne Université, Inserm, Paris, France
| | - Anatol Kontush
- ICAN, National Institute for Health and Medical Research, Paris, France
| | - Damarys Loew
- CurieCoreTech Spectrométrie de Masse Protéomique, Institut Curie, PSL Research University, Paris, France
| | - Marie Lhomme
- Foundation for Innovation in Cardiometabolism and Nutrition (IHU ICAN, ICAN OMICS and ICAN I/O), F-75013, Paris, France
| | - Wouter H Roos
- Moleculaire Biofysica, Zernike Instituut, Rijksuniversiteit Groningen, Groningen, The Netherlands
| | - Graça Raposo
- Institut Curie, PSL Research University, CNRS, UMR144, 26 rue d'Ulm, 75248, Paris Cedex 05, France
- Institut Curie, PSL Research University, CNRS, UMR144, Cell and Tissue Imaging Facility (PICT-IBiSA), 26, rue d'Ulm, 75248, Paris Cedex 05, France
| | - Guillaume van Niel
- Institut Curie, PSL Research University, CNRS, UMR144, 26 rue d'Ulm, 75248, Paris Cedex 05, France.
- Institute of Psychiatry and Neuroscience of Paris (IPNP), Université de Paris, Paris, France.
- CRCI2NA, Nantes Université, Inserm UMR1307, CNRS UMR6075, Université d'Angers, Nantes, France.
- GHU-Paris Psychiatrie et Neurosciences, Hôpital Sainte Anne, Paris, France.
| |
Collapse
|
24
|
Cho Y, Cho MY, Yoon J, Hong DE, Lee J, Park HS, Lee H, Hong KS, Won‐Kyu L, Saehae C, Song S, Noh Y. Evaluation of unmodified human cell-derived extracellular vesicle mitochondrial deoxyribonucleic acid-based biodistribution in rodents. J Extracell Vesicles 2024; 13:e12489. [PMID: 39016198 PMCID: PMC11253025 DOI: 10.1002/jev2.12489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Revised: 06/10/2024] [Accepted: 07/02/2024] [Indexed: 07/18/2024] Open
Abstract
Recently, extracellular vesicles (EVs) have been developed as therapeutic targets for various diseases. Biodistribution is crucial for EVs intended for therapeutic purposes because it can determine the degree of on- and off-target effects. This study aimed to explore techniques to evaluate the biodistribution of unmodified EVs. We devised a novel quantitative polymerase chain reaction (qPCR)-based assay to detect unmodified EVs by targeting mitochondrial deoxyribonucleic acid (mtDNA), a constituent of EVs. We focused on specific mtDNA regions that exhibited homologous variations distinct from their rodent mtDNA counterparts to establish this analytical approach. Herein, we successfully designed primers and probes targeting human and rodent mtDNA sequences and developed a highly specific and sensitive qPCR method. Furthermore, the quantification range of EVs isolated from various cells differed based on the manufacturer and cell source. IRDye 800CW-labelled Expi293F EV mimetics were administered to the animals via the tail vein to compare the imaging test and mtDNA-qPCR results. The results obtained from imaging tests and mtDNA-qPCR to investigate EV biodistribution patterns revealed differences. The results revealed that our newly developed method effectively determined the biodistribution of unmodified EVs with high sensitivity and reproducibility.
Collapse
Affiliation(s)
- Young‐Woo Cho
- Division of Drug Safety EvaluationNDDC, Osong Medical Innovation FoundationCheongjuSouth Korea
- College of PharmacyChungbuk National UniversityCheongjuSouth Korea
| | - Mi Young Cho
- Biopharmaceutical Research CenterKorea Basic Science InstituteCheongjuSouth Korea
| | - Jaehyeon Yoon
- Division of Drug Safety EvaluationNDDC, Osong Medical Innovation FoundationCheongjuSouth Korea
| | - Da Eun Hong
- Division of Drug Safety EvaluationNDDC, Osong Medical Innovation FoundationCheongjuSouth Korea
| | - Ju‐young Lee
- Division of Drug Safety EvaluationNDDC, Osong Medical Innovation FoundationCheongjuSouth Korea
- College of PharmacyChungbuk National UniversityCheongjuSouth Korea
| | - Hye Sun Park
- Biopharmaceutical Research CenterKorea Basic Science InstituteCheongjuSouth Korea
| | - Hyunseung Lee
- Biopharmaceutical Research CenterKorea Basic Science InstituteCheongjuSouth Korea
| | - Kwan Soo Hong
- Biopharmaceutical Research CenterKorea Basic Science InstituteCheongjuSouth Korea
- Department of ChemistryChung‐Ang UniversitySeoulSouth Korea
| | - Lee Won‐Kyu
- Division of Drug Safety EvaluationNDDC, Osong Medical Innovation FoundationCheongjuSouth Korea
| | - Choi Saehae
- Division of Drug Safety EvaluationNDDC, Osong Medical Innovation FoundationCheongjuSouth Korea
| | - Suk‐Gil Song
- College of PharmacyChungbuk National UniversityCheongjuSouth Korea
| | - Young‐Woock Noh
- Division of Drug Safety EvaluationNDDC, Osong Medical Innovation FoundationCheongjuSouth Korea
| |
Collapse
|
25
|
Tang X, He Y, Liu J, Xu J, Peng Q. Exosomes: The endogenous nanomaterials packed with potential for diagnosis and treatment of neurologic disorders. Colloids Surf B Biointerfaces 2024; 239:113938. [PMID: 38718474 DOI: 10.1016/j.colsurfb.2024.113938] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 04/14/2024] [Accepted: 04/30/2024] [Indexed: 03/17/2025]
Abstract
Neurologic disorders (NDs) are serious diseases that threaten public health. However, due to the complex pathogenesis and significant individual differences in traditional treatments, specific treatment methods for NDs are still lacking. Exosomes, the smallest extracellular vesicles secreted by eukaryotic cells, are receiving increasing attention in the field of NDs. They contain misfolded proteins related to various NDs, including amyloid-beta, Tau proteins, and α-synuclein, indicating their promising roles in the diagnosis and treatment of NDs. In this review, an overview of the biogenesis, composition, and biological functions of exosomes is provided. Moreover, we summarize their potential roles in the pathogenesis of three prevalent NDs (including Alzheimer's disease, Ischemic stroke, and Parkinson's disease). On this basis, the diagnostic potential and therapeutic value of exosomes carrying various bioactive molecules are discussed in detail. Also, the concerns and perspectives of exosome-based diagnosis and therapy are discussed.
Collapse
Affiliation(s)
- Xuelin Tang
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yuxuan He
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Jinchi Liu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Jingchen Xu
- Department of Dental Medical Center, China-Japan Friendship Hospital, Beijing 100029, China.
| | - Qiang Peng
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
26
|
Farahzadi R, Fathi E, Valipour B, Ghaffary S. Stem cells-derived exosomes as cardiac regenerative agents. IJC HEART & VASCULATURE 2024; 52:101399. [PMID: 38584674 PMCID: PMC10990901 DOI: 10.1016/j.ijcha.2024.101399] [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: 12/07/2023] [Revised: 03/03/2024] [Accepted: 03/28/2024] [Indexed: 04/09/2024]
Abstract
Heart failure is a root cause of morbidity and mortality worldwide. Due to the limited regenerative capacity of the heart following myocardial injury, stem cell-based therapies have been considered a hopeful approach for improving cardiac regeneration. In recent years, different kinds of cell products have been investigated regarding their potential to treat patients with heart failure. Despite special attention to cell therapy and its products, therapeutic efficacy has been disappointing, and clinical application is not affordable. In the past few years, a subset of small extracellular vehicles (EVs), commonly known as "exosomes," was reported to grant regenerative and cardioprotective signals at a value similar to their donor cells. The conceptual advantage is that they may be ideally used without evoking a relevant recipient immune response or other adverse effects associated with viable cells. The evidence related to their beneficial effects in animal models of heart failure is rapidly growing. However, there is remarkable heterogeneity regarding source cells, isolation process, effective dosage, and delivery mode. This brief review will focus on the latest research and debates on regenerative potential and cardiac repair of exosomes from different sources, such as cardiac/non-cardiac stem, somatic cells, and progenitor cells. Overall, the current state of research on exosomes as an experimental therapy for heart diseases will be discussed.
Collapse
Affiliation(s)
- Raheleh Farahzadi
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Medical Philosophy and History Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ezzatollah Fathi
- Department of Clinical Sciences, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran
| | - Behnaz Valipour
- Department of Anatomical Sciences, Sarab Faculty of Medical Sciences, Sarab, Iran
- Department of Anatomical Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Saba Ghaffary
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
27
|
Baginska U, Balagura G, Toonen RF, Verhage M. High-throughput assay for regulated secretion of neuropeptides in mouse and human neurons. J Biol Chem 2024; 300:107321. [PMID: 38677517 PMCID: PMC11170154 DOI: 10.1016/j.jbc.2024.107321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 04/17/2024] [Accepted: 04/20/2024] [Indexed: 04/29/2024] Open
Abstract
Neuropeptides are the largest group of chemical signals in the brain. More than 100 different neuropeptides modulate various brain functions and their dysregulation has been associated with neurological disorders. Neuropeptides are packed into dense core vesicles (DCVs), which fuse with the plasma membrane in a calcium-dependent manner. Here, we describe a novel high-throughput assay for DCV exocytosis using a chimera of Nanoluc luciferase and the DCV-cargo neuropeptide Y (NPY). The NPY-Nanoluc reporter colocalized with endogenous DCV markers in all neurons with little mislocalization to other cellular compartments. NPY-Nanoluc reported DCV exocytosis in both rodent and induced pluripotent stem cell-derived human neurons, with similar depolarization, Ca2+, RAB3, and STXBP1/MUNC18 dependence as low-throughput assays. Moreover, NPY-Nanoluc accurately reported modulation of DCV exocytosis by known modulators diacylglycerol analog and Ca2+ channel blocker and showed a higher assay sensitivity than a widely used single-cell low-throughput assay. Lastly, we showed that Nanoluc coupled to other secretory markers reports on constitutive secretion. In conclusion, the NPY-Nanoluc is a sensitive reporter of DCV exocytosis in mammalian neurons, suitable for pharmacological and genomic screening for DCV exocytosis genes and for mechanism-based treatments for central nervous system disorders.
Collapse
Affiliation(s)
- Urszula Baginska
- Department of Functional Genomics, Faculty of Exact Science, Center for Neurogenomics and Cognitive Research, VU University Amsterdam and VU Medical Center, Amsterdam, The Netherlands
| | - Ganna Balagura
- Department of Functional Genomics, Faculty of Exact Science, Center for Neurogenomics and Cognitive Research, VU University Amsterdam and VU Medical Center, Amsterdam, The Netherlands; Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, and Maternal and Child Health, University of Genoa, Genoa, Italy; Pediatric Neurology and Muscular Diseases Unit, IRCCS 'G. Gaslini' Institute, Genoa, Italy
| | - Ruud F Toonen
- Department of Functional Genomics, Faculty of Exact Science, Center for Neurogenomics and Cognitive Research, VU University Amsterdam and VU Medical Center, Amsterdam, The Netherlands
| | - Matthijs Verhage
- Department of Functional Genomics, Faculty of Exact Science, Center for Neurogenomics and Cognitive Research, VU University Amsterdam and VU Medical Center, Amsterdam, The Netherlands; Human Genetics, Amsterdam University Medical Center, Amsterdam, The Netherlands.
| |
Collapse
|
28
|
Zhang B, Lai RC, Sim WK, Tan TT, Lim SK. An Assessment of Administration Route on MSC-sEV Therapeutic Efficacy. Biomolecules 2024; 14:622. [PMID: 38927026 PMCID: PMC11202284 DOI: 10.3390/biom14060622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 05/15/2024] [Accepted: 05/22/2024] [Indexed: 06/28/2024] Open
Abstract
Mesenchymal stem/stromal cell-derived small extracellular vesicles (MSC-sEVs) are promising therapeutic agents. In this study, we investigated how the administration route of MSC-sEVs affects their therapeutic efficacy in a mouse model of bleomycin (BLM)-induced skin scleroderma (SSc). We evaluated the impact of topical (TOP), subcutaneous (SC), and intraperitoneal (IP) administration of MSC-sEVs on dermal fibrosis, collagen density, and thickness. All three routes of administration significantly reduced BLM-induced fibrosis in the skin, as determined by Masson's Trichrome staining. However, only TOP administration reduced BLM-induced dermal collagen density, with no effect on dermal thickness observed for all administration routes. Moreover, SC, but not TOP or IP administration, increased anti-inflammatory profibrotic CD163+ M2 macrophages. These findings indicate that the administration route influences the therapeutic efficacy of MSC-sEVs in alleviating dermal fibrosis, with TOP administration being the most effective, and this efficacy is not mediated by M2 macrophages. Since both TOP and SC administration target the skin, the difference in their efficacy likely stems from variations in MSC-sEV delivery in the skin. Fluorescence-labelled TOP, but not SC MSC-sEVs when applied to skin explant cultures, localized in the stratum corneum. Hence, the superior efficacy of TOP over SC MSC-sEVs could be attributed to this localization. A comparison of the proteomes of stratum corneum and MSC-sEVs revealed the presence of >100 common proteins. Most of these proteins, such as filaggrin, were known to be crucial for maintaining skin barrier function against irritants and toxins, thereby mitigating inflammation-induced fibrosis. Therefore, the superior efficacy of TOP MSC-sEVs over SC and IP MSC-sEVs against SSc is mediated by the delivery of proteins to the stratum corneum to reinforce the skin barrier.
Collapse
Affiliation(s)
- Bin Zhang
- Paracrine Therapeutics Pte. Ltd., 1 Tai Seng Ave, #02-04 Tai Seng Exchange, Singapore 536464, Singapore; (B.Z.); (R.C.L.); (W.K.S.); (T.T.T.)
| | - Ruenn Chai Lai
- Paracrine Therapeutics Pte. Ltd., 1 Tai Seng Ave, #02-04 Tai Seng Exchange, Singapore 536464, Singapore; (B.Z.); (R.C.L.); (W.K.S.); (T.T.T.)
| | - Wei Kian Sim
- Paracrine Therapeutics Pte. Ltd., 1 Tai Seng Ave, #02-04 Tai Seng Exchange, Singapore 536464, Singapore; (B.Z.); (R.C.L.); (W.K.S.); (T.T.T.)
| | - Thong Teck Tan
- Paracrine Therapeutics Pte. Ltd., 1 Tai Seng Ave, #02-04 Tai Seng Exchange, Singapore 536464, Singapore; (B.Z.); (R.C.L.); (W.K.S.); (T.T.T.)
| | - Sai Kiang Lim
- Paracrine Therapeutics Pte. Ltd., 1 Tai Seng Ave, #02-04 Tai Seng Exchange, Singapore 536464, Singapore; (B.Z.); (R.C.L.); (W.K.S.); (T.T.T.)
- Department of Surgery, YLL School of Medicine, National University Singapore (NUS), 5 Lower Kent Ridge Road, Singapore 119074, Singapore
| |
Collapse
|
29
|
Liu M, Zhang Y, He J, Liu W, Li Z, Zhang Y, Gu A, Zhao M, Liu M, Liu X. Fusion with ARRDC1 or CD63: A Strategy to Enhance p53 Loading into Extracellular Vesicles for Tumor Suppression. Biomolecules 2024; 14:591. [PMID: 38785998 PMCID: PMC11118238 DOI: 10.3390/biom14050591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 05/04/2024] [Accepted: 05/14/2024] [Indexed: 05/25/2024] Open
Abstract
Small extracellular vesicles (sEVs) have emerged as promising therapeutic agents and drug delivery vehicles. Targeted modification of sEVs and their contents using genetic modification strategies is one of the most popular methods. This study investigated the effects of p53 fusion with arrestin domain-containing protein 1 (ARRDC1) and CD63 on the generation of sEVs, p53 loading efficiency, and therapeutic efficacy. Overexpression of either ARRDC1-p53 (ARP) or CD63-p53 (CDP) significantly elevated p53 mRNA and protein levels. The incorporation of ARRDC1 and CD63 significantly enhanced HEK293T-sEV biogenesis, evidenced by significant increases in sEV-associated proteins TSG101 and LAMP1, resulting in a boost in sEV production. Importantly, fusion with ARRDC1 or CD63 substantially increased the efficiency of loading both p53 fusion proteins and its mRNA into sEVs. sEVs equipped with ARP or CDP significantly enhanced the enrichment of p53 fusion proteins and mRNA in p53-null H1299 cells, resulting in a marked increase in apoptosis and a reduction in cell proliferation, with ARP-sEVs demonstrating greater effectiveness than CDP-sEVs. These findings underscore the enhanced functionality of ARRDC1- and CD63-modified sEVs, emphasizing the potential of genetic modifications in sEV-based therapies for targeted cancer treatment.
Collapse
Affiliation(s)
- Min Liu
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha 410078, China; (M.L.); (Y.Z.); (J.H.); (W.L.); (Z.L.); (Y.Z.); (A.G.); (M.Z.)
| | - Yu Zhang
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha 410078, China; (M.L.); (Y.Z.); (J.H.); (W.L.); (Z.L.); (Y.Z.); (A.G.); (M.Z.)
| | - Jianfeng He
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha 410078, China; (M.L.); (Y.Z.); (J.H.); (W.L.); (Z.L.); (Y.Z.); (A.G.); (M.Z.)
- Department of Cell Biology, School of Life Sciences, Central South University, Changsha 410013, China
| | - Wanxi Liu
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha 410078, China; (M.L.); (Y.Z.); (J.H.); (W.L.); (Z.L.); (Y.Z.); (A.G.); (M.Z.)
| | - Zhexuan Li
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha 410078, China; (M.L.); (Y.Z.); (J.H.); (W.L.); (Z.L.); (Y.Z.); (A.G.); (M.Z.)
| | - Yiti Zhang
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha 410078, China; (M.L.); (Y.Z.); (J.H.); (W.L.); (Z.L.); (Y.Z.); (A.G.); (M.Z.)
| | - Ao Gu
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha 410078, China; (M.L.); (Y.Z.); (J.H.); (W.L.); (Z.L.); (Y.Z.); (A.G.); (M.Z.)
| | - Mingri Zhao
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha 410078, China; (M.L.); (Y.Z.); (J.H.); (W.L.); (Z.L.); (Y.Z.); (A.G.); (M.Z.)
| | - Mujun Liu
- Department of Cell Biology, School of Life Sciences, Central South University, Changsha 410013, China
- Hunan Key Laboratory of Basic and Applied Hematology, Central South University, Changsha 410078, China
- Hunan Key Laboratory of Animal Models for Human Diseases, Central South University, Changsha 410078, China
| | - Xionghao Liu
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha 410078, China; (M.L.); (Y.Z.); (J.H.); (W.L.); (Z.L.); (Y.Z.); (A.G.); (M.Z.)
- Hunan Key Laboratory of Basic and Applied Hematology, Central South University, Changsha 410078, China
- Hunan Key Laboratory of Animal Models for Human Diseases, Central South University, Changsha 410078, China
| |
Collapse
|
30
|
Choi W, Park DJ, Eliceiri BP. Defining tropism and activity of natural and engineered extracellular vesicles. Front Immunol 2024; 15:1363185. [PMID: 38660297 PMCID: PMC11039936 DOI: 10.3389/fimmu.2024.1363185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Accepted: 03/25/2024] [Indexed: 04/26/2024] Open
Abstract
Extracellular vesicles (EVs) have important roles as mediators of cell-to-cell communication, with physiological functions demonstrated in various in vivo models. Despite advances in our understanding of the biological function of EVs and their potential for use as therapeutics, there are limitations to the clinical approaches for which EVs would be effective. A primary determinant of the biodistribution of EVs is the profile of proteins and other factors on the surface of EVs that define the tropism of EVs in vivo. For example, proteins displayed on the surface of EVs can vary in composition by cell source of the EVs and the microenvironment into which EVs are delivered. In addition, interactions between EVs and recipient cells that determine uptake and endosomal escape in recipient cells affect overall systemic biodistribution. In this review, we discuss the contribution of the EV donor cell and the role of the microenvironment in determining EV tropism and thereby determining the uptake and biological activity of EVs.
Collapse
Affiliation(s)
- Wooil Choi
- Department of Surgery, University of California San Diego, La Jolla, CA, United States
| | - Dong Jun Park
- Department of Surgery, University of California San Diego, La Jolla, CA, United States
| | - Brian P. Eliceiri
- Department of Surgery, University of California San Diego, La Jolla, CA, United States
- Department of Dermatology, University of California San Diego, La Jolla, CA, United States
| |
Collapse
|
31
|
Creeden JF, Sevier J, Zhang JT, Lapitsky Y, Brunicardi FC, Jin G, Nemunaitis J, Liu JY, Kalinoski A, Rao D, Liu SH. Smart exosomes enhance PDAC targeted therapy. J Control Release 2024; 368:413-429. [PMID: 38431093 DOI: 10.1016/j.jconrel.2024.02.037] [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: 11/02/2023] [Revised: 02/21/2024] [Accepted: 02/25/2024] [Indexed: 03/05/2024]
Abstract
Exosomes continue to attract interest as a promising nanocarrier drug delivery technology. They are naturally derived nanoscale extracellular vesicles with innate properties well suited to shuttle proteins, lipids, and nucleic acids between cells. Nonetheless, their clinical utility is currently limited by several major challenges, such as their inability to target tumor cells and a high proportion of clearance by the mononuclear phagocyte system (MPS) of the liver and spleen. To overcome these limitations, we developed "Smart Exosomes" that co-display RGD and CD47p110-130 through CD9 engineering (ExoSmart). The resultant ExoSmart demonstrates enhanced binding capacity to αvβ3 on pancreatic ductal adenocarcinoma (PDAC) cells, resulting in amplified cellular uptake in in vitro and in vivo models and increased chemotherapeutic efficacies. Simultaneously, ExoSmart significantly reduced liver and spleen clearance of exosomes by inhibiting macrophage phagocytosis via CD47p110-130 interaction with signal regulatory proteins (SIRPα) on macrophages. These studies demonstrate that an engineered exosome drug delivery system increases PDAC therapeutic efficacy by enhancing active PDAC targeting and prolonging circulation times, and their findings hold tremendous translational potential for cancer therapy while providing a concrete foundation for future work utilizing novel peptide-engineered exosome strategies.
Collapse
Affiliation(s)
- Justin F Creeden
- Department of Cell and Cancer Biology, University of Toledo, Toledo, OH, USA
| | - Jonathan Sevier
- Department of Cell and Cancer Biology, University of Toledo, Toledo, OH, USA
| | - Jian-Ting Zhang
- Department of Cell and Cancer Biology, University of Toledo, Toledo, OH, USA
| | - Yakov Lapitsky
- Department of Chemical Engineering, University of Toledo, Toledo, OH, USA
| | - F Charles Brunicardi
- Department of Surgery, SUNY Downstate Health Sciences University, Brooklyn, NY, USA
| | - Ge Jin
- Department of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | | | - Jing-Yuan Liu
- Department of Medicine, University of Toledo, Toledo, OH, USA
| | | | | | - Shi-He Liu
- Department of Cell and Cancer Biology, University of Toledo, Toledo, OH, USA.
| |
Collapse
|
32
|
Boudna M, Campos AD, Vychytilova-Faltejskova P, Machackova T, Slaby O, Souckova K. Strategies for labelling of exogenous and endogenous extracellular vesicles and their application for in vitro and in vivo functional studies. Cell Commun Signal 2024; 22:171. [PMID: 38461237 PMCID: PMC10924393 DOI: 10.1186/s12964-024-01548-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Accepted: 02/29/2024] [Indexed: 03/11/2024] Open
Abstract
This review presents a comprehensive overview of labelling strategies for endogenous and exogenous extracellular vesicles, that can be utilised both in vitro and in vivo. It covers a broad spectrum of approaches, including fluorescent and bioluminescent labelling, and provides an analysis of their applications, strengths, and limitations. Furthermore, this article presents techniques that use radioactive tracers and contrast agents with the ability to track EVs both spatially and temporally. Emphasis is also placed on endogenous labelling mechanisms, represented by Cre-lox and CRISPR-Cas systems, which are powerful and flexible tools for real-time EV monitoring or tracking their fate in target cells. By summarizing the latest developments across these diverse labelling techniques, this review provides researchers with a reference to select the most appropriate labelling method for their EV based research.
Collapse
Affiliation(s)
- Marie Boudna
- Central European Institute of Technology, Masaryk University, Kamenice 753/5, 625 00, Brno, Czech Republic
- Department of Biology, Faculty of Medicine, Masaryk University, Kamenice 753/5, 625 00, Brno, Czech Republic
| | - Andres Delgado Campos
- Central European Institute of Technology, Masaryk University, Kamenice 753/5, 625 00, Brno, Czech Republic
| | | | - Tana Machackova
- Central European Institute of Technology, Masaryk University, Kamenice 753/5, 625 00, Brno, Czech Republic
| | - Ondrej Slaby
- Central European Institute of Technology, Masaryk University, Kamenice 753/5, 625 00, Brno, Czech Republic.
- Department of Biology, Faculty of Medicine, Masaryk University, Kamenice 753/5, 625 00, Brno, Czech Republic.
| | - Kamila Souckova
- Central European Institute of Technology, Masaryk University, Kamenice 753/5, 625 00, Brno, Czech Republic.
| |
Collapse
|
33
|
Nannan L, Decombis S, Terryn C, Audonnet S, Michel J, Brassart‐Pasco S, Gsell W, Himmelreich U, Brassart B. Dysregulation of intercellular communication in vitro and in vivo via extracellular vesicles secreted by pancreatic duct adenocarcinoma cells and generated under the influence of the AG9 elastin peptide-conditioned microenvironment. JOURNAL OF EXTRACELLULAR BIOLOGY 2024; 3:e145. [PMID: 38939412 PMCID: PMC11080898 DOI: 10.1002/jex2.145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Revised: 01/24/2024] [Accepted: 02/22/2024] [Indexed: 06/29/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an aggressive malignancy with poor prognosis due to its highly metastatic profile. Intercellular communication between cancer and stromal cells via extracellular vesicles (EVs) is crucial for the premetastatic microenvironment preparation leading to tumour metastasis. This study shows that under the influence of bioactive peptides derived from the extracellular matrix microenvironment, illustrated here by the AG-9 elastin-derived peptide (EDP), PDAC cells secrete more tumour-derived EVs. Compared to PDAC-derived EVs, tumour-derived EVs resulting from AG-9 treatment (PDAC AG-9-derived EVs) significantly stimulated cell proliferation. At constant amount, tumour-derived EVs were similarly taken up by PDAC and HMEC-1 cells. Tumour-derived EVs stimulated cell proliferation, migration, proteinase secretion, and angiogenesis. Bioluminescence imaging allowed tumour-derived EV/FLuc+ tracking in vivo in a PDAC mouse model. The biodistribution of PDAC AG-9-derived EVs was different to PDAC-derived EVs. Our results demonstrate that the microenvironment, through EDP release, may not only influence the genesis of EVs but may also affect tumour progression (tumour growth and angiogenesis), and metastatic homing by modifying the in vivo biodistribution of tumour-derived EVs. They are potential candidates for targeted drug delivery and modulation of tumour progression, and they constitute a new generation of therapeutic tools, merging oncology and genic therapy.
Collapse
Affiliation(s)
- Lise Nannan
- Université de Reims Champagne‐ArdenneLaboratoire de Biochimie Médicale et Biologie Moléculaire, UFR MédecineReimsFrance
- CNRS UMR 7369 Matrice Extracellulaire et Dynamique CellulaireReimsFrance
- KU Leuven, Department of Imaging and PathologyBiomedical MRILeuvenBelgium
| | - Salomé Decombis
- Université de Reims Champagne‐ArdenneLaboratoire de Biochimie Médicale et Biologie Moléculaire, UFR MédecineReimsFrance
- CNRS UMR 7369 Matrice Extracellulaire et Dynamique CellulaireReimsFrance
| | | | - Sandra Audonnet
- URCACyt PlatformUniversity of Reims Champagne‐ArdenneReimsFrance
| | - Jean Michel
- Inserm, Université de Reims Champagne‐Ardenne, P3Cell UMR‐S1250, SFR CAP‐SANTEReimsFrance
| | - Sylvie Brassart‐Pasco
- Université de Reims Champagne‐ArdenneLaboratoire de Biochimie Médicale et Biologie Moléculaire, UFR MédecineReimsFrance
- CNRS UMR 7369 Matrice Extracellulaire et Dynamique CellulaireReimsFrance
| | - Willy Gsell
- KU Leuven, Department of Imaging and PathologyBiomedical MRILeuvenBelgium
| | - Uwe Himmelreich
- KU Leuven, Department of Imaging and PathologyBiomedical MRILeuvenBelgium
| | - Bertrand Brassart
- Université de Reims Champagne‐ArdenneLaboratoire de Biochimie Médicale et Biologie Moléculaire, UFR MédecineReimsFrance
- CNRS UMR 7369 Matrice Extracellulaire et Dynamique CellulaireReimsFrance
| |
Collapse
|
34
|
Vaswani CM, Simone J, Pavelick JL, Wu X, Tan GW, Ektesabi AM, Gupta S, Tsoporis JN, Dos Santos CC. Tiny Guides, Big Impact: Focus on the Opportunities and Challenges of miR-Based Treatments for ARDS. Int J Mol Sci 2024; 25:2812. [PMID: 38474059 DOI: 10.3390/ijms25052812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Revised: 02/24/2024] [Accepted: 02/25/2024] [Indexed: 03/14/2024] Open
Abstract
Acute Respiratory Distress Syndrome (ARDS) is characterized by lung inflammation and increased membrane permeability, which represents the leading cause of mortality in ICUs. Mechanical ventilation strategies are at the forefront of supportive approaches for ARDS. Recently, an increasing understanding of RNA biology, function, and regulation, as well as the success of RNA vaccines, has spurred enthusiasm for the emergence of novel RNA-based therapeutics. The most common types of RNA seen in development are silencing (si)RNAs, antisense oligonucleotide therapy (ASO), and messenger (m)RNAs that collectively account for 80% of the RNA therapeutics pipeline. These three RNA platforms are the most mature, with approved products and demonstrated commercial success. Most recently, miRNAs have emerged as pivotal regulators of gene expression. Their dysregulation in various clinical conditions offers insights into ARDS pathogenesis and offers the innovative possibility of using microRNAs as targeted therapy. This review synthesizes the current state of the literature to contextualize the therapeutic potential of miRNA modulation. It considers the potential for miR-based therapeutics as a nuanced approach that incorporates the complexity of ARDS pathophysiology and the multifaceted nature of miRNA interactions.
Collapse
Affiliation(s)
- Chirag M Vaswani
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, University of Toronto, Toronto, ON M5B 1W8, Canada
| | - Julia Simone
- Department of Medicine, McMaster University, Hamilton, ON L8V 5C2, Canada
| | - Jacqueline L Pavelick
- Institute of Medical Sciences, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Xiao Wu
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, University of Toronto, Toronto, ON M5B 1W8, Canada
| | - Greaton W Tan
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, University of Toronto, Toronto, ON M5B 1W8, Canada
| | - Amin M Ektesabi
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, University of Toronto, Toronto, ON M5B 1W8, Canada
- Institute of Medical Sciences, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Sahil Gupta
- Faculty of Medicine, School of Medicine, The University of Queensland, Herston, QLD 4006, Australia
| | - James N Tsoporis
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, University of Toronto, Toronto, ON M5B 1W8, Canada
| | - Claudia C Dos Santos
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, University of Toronto, Toronto, ON M5B 1W8, Canada
- Institute of Medical Sciences, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
- Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
- Interdepartmental Division of Critical Care, St. Michael's Hospital, University of Toronto, Toronto, ON M5B 1W8, Canada
| |
Collapse
|
35
|
Jackson HK, Long HM, Yam‐Puc JC, Palmulli R, Haigh TA, Gerber PP, Lee JS, Matheson NJ, Young L, Trowsdale J, Lo M, Taylor GS, Thaventhiran JE, Edgar JR. Bioengineered small extracellular vesicles deliver multiple SARS-CoV-2 antigenic fragments and drive a broad immunological response. J Extracell Vesicles 2024; 13:e12412. [PMID: 38339765 PMCID: PMC10858312 DOI: 10.1002/jev2.12412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 12/22/2023] [Accepted: 01/23/2024] [Indexed: 02/12/2024] Open
Abstract
The COVID-19 pandemic highlighted the clear risk that zoonotic viruses pose to global health and economies. The scientific community responded by developing several efficacious vaccines which were expedited by the global need for vaccines. The emergence of SARS-CoV-2 breakthrough infections highlights the need for additional vaccine modalities to provide stronger, long-lived protective immunity. Here we report the design and preclinical testing of small extracellular vesicles (sEVs) as a multi-subunit vaccine. Cell lines were engineered to produce sEVs containing either the SARS-CoV-2 Spike receptor-binding domain, or an antigenic region from SARS-CoV-2 Nucleocapsid, or both in combination, and we tested their ability to evoke immune responses in vitro and in vivo. B cells incubated with bioengineered sEVs were potent activators of antigen-specific T cell clones. Mice immunised with sEVs containing both sRBD and Nucleocapsid antigens generated sRBD-specific IgGs, nucleocapsid-specific IgGs, which neutralised SARS-CoV-2 infection. sEV-based vaccines allow multiple antigens to be delivered simultaneously resulting in potent, broad immunity, and provide a quick, cheap, and reliable method to test vaccine candidates.
Collapse
Affiliation(s)
- Hannah K. Jackson
- Department of PathologyUniversity of CambridgeCambridgeUK
- Exosis, Inc. Palm BeachPalm BeachFloridaUSA
| | - Heather M. Long
- Institute of Immunology and ImmunotherapyUniversity of BirminghamBirminghamUK
| | | | | | - Tracey A. Haigh
- Institute of Immunology and ImmunotherapyUniversity of BirminghamBirminghamUK
| | - Pehuén Pereyra Gerber
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID)University of CambridgeCambridgeUK
- Department of MedicineUniversity of CambridgeCambridgeUK
| | - Jin S. Lee
- Department of PathologyUniversity of CambridgeCambridgeUK
| | - Nicholas J. Matheson
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID)University of CambridgeCambridgeUK
- Department of MedicineUniversity of CambridgeCambridgeUK
- NHS Blood and TransplantCambridgeUK
| | | | | | - Mathew Lo
- Exosis, Inc. Palm BeachPalm BeachFloridaUSA
| | - Graham S. Taylor
- Institute of Immunology and ImmunotherapyUniversity of BirminghamBirminghamUK
| | | | - James R. Edgar
- Department of PathologyUniversity of CambridgeCambridgeUK
| |
Collapse
|
36
|
Hermann DM, Peruzzotti-Jametti L, Giebel B, Pluchino S. Extracellular vesicles set the stage for brain plasticity and recovery by multimodal signalling. Brain 2024; 147:372-389. [PMID: 37768167 PMCID: PMC10834259 DOI: 10.1093/brain/awad332] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 08/07/2023] [Accepted: 09/15/2023] [Indexed: 09/29/2023] Open
Abstract
Extracellular vesicles (EVs) are extremely versatile naturally occurring membrane particles that convey complex signals between cells. EVs of different cellular sources are capable of inducing striking therapeutic responses in neurological disease models. Differently from pharmacological compounds that act by modulating defined signalling pathways, EV-based therapeutics possess multiple abilities via a variety of effectors, thus allowing the modulation of complex disease processes that may have very potent effects on brain tissue recovery. When applied in vivo in experimental models of neurological diseases, EV-based therapeutics have revealed remarkable effects on immune responses, cell metabolism and neuronal plasticity. This multimodal modulation of neuroimmune networks by EVs profoundly influences disease processes in a highly synergistic and context-dependent way. Ultimately, the EV-mediated restoration of cellular functions helps to set the stage for neurological recovery. With this review we first outline the current understanding of the mechanisms of action of EVs, describing how EVs released from various cellular sources identify their cellular targets and convey signals to recipient cells. Then, mechanisms of action applicable to key neurological conditions such as stroke, multiple sclerosis and neurodegenerative diseases are presented. Pathways that deserve attention in specific disease contexts are discussed. We subsequently showcase considerations about EV biodistribution and delineate genetic engineering strategies aiming at enhancing brain uptake and signalling. By sketching a broad view of EV-orchestrated brain plasticity and recovery, we finally define possible future clinical EV applications and propose necessary information to be provided ahead of clinical trials. Our goal is to provide a steppingstone that can be used to critically discuss EVs as next generation therapeutics for brain diseases.
Collapse
Affiliation(s)
- Dirk M Hermann
- Department of Neurology, University Hospital Essen, University of Duisburg-Essen, D-45122 Essen, Germany
| | - Luca Peruzzotti-Jametti
- Department of Clinical Neurosciences and National Institute for Health Research (NIHR) Biomedical Research Centre, University of Cambridge, Cambridge CB2 0AH, UK
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London W12 0NN, UK
| | - Bernd Giebel
- Institute of Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, D-45147 Essen, Germany
| | - Stefano Pluchino
- Department of Clinical Neurosciences and National Institute for Health Research (NIHR) Biomedical Research Centre, University of Cambridge, Cambridge CB2 0AH, UK
| |
Collapse
|
37
|
Welsh JA, Goberdhan DCI, O'Driscoll L, Buzas EI, Blenkiron C, Bussolati B, Cai H, Di Vizio D, Driedonks TAP, Erdbrügger U, Falcon‐Perez JM, Fu Q, Hill AF, Lenassi M, Lim SK, Mahoney MG, Mohanty S, Möller A, Nieuwland R, Ochiya T, Sahoo S, Torrecilhas AC, Zheng L, Zijlstra A, Abuelreich S, Bagabas R, Bergese P, Bridges EM, Brucale M, Burger D, Carney RP, Cocucci E, Colombo F, Crescitelli R, Hanser E, Harris AL, Haughey NJ, Hendrix A, Ivanov AR, Jovanovic‐Talisman T, Kruh‐Garcia NA, Ku'ulei‐Lyn Faustino V, Kyburz D, Lässer C, Lennon KM, Lötvall J, Maddox AL, Martens‐Uzunova ES, Mizenko RR, Newman LA, Ridolfi A, Rohde E, Rojalin T, Rowland A, Saftics A, Sandau US, Saugstad JA, Shekari F, Swift S, Ter‐Ovanesyan D, Tosar JP, Useckaite Z, Valle F, Varga Z, van der Pol E, van Herwijnen MJC, Wauben MHM, Wehman AM, Williams S, Zendrini A, Zimmerman AJ, MISEV Consortium, Théry C, Witwer KW. Minimal information for studies of extracellular vesicles (MISEV2023): From basic to advanced approaches. J Extracell Vesicles 2024; 13:e12404. [PMID: 38326288 PMCID: PMC10850029 DOI: 10.1002/jev2.12404] [Citation(s) in RCA: 1038] [Impact Index Per Article: 1038.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 12/15/2023] [Accepted: 12/19/2023] [Indexed: 02/09/2024] Open
Abstract
Extracellular vesicles (EVs), through their complex cargo, can reflect the state of their cell of origin and change the functions and phenotypes of other cells. These features indicate strong biomarker and therapeutic potential and have generated broad interest, as evidenced by the steady year-on-year increase in the numbers of scientific publications about EVs. Important advances have been made in EV metrology and in understanding and applying EV biology. However, hurdles remain to realising the potential of EVs in domains ranging from basic biology to clinical applications due to challenges in EV nomenclature, separation from non-vesicular extracellular particles, characterisation and functional studies. To address the challenges and opportunities in this rapidly evolving field, the International Society for Extracellular Vesicles (ISEV) updates its 'Minimal Information for Studies of Extracellular Vesicles', which was first published in 2014 and then in 2018 as MISEV2014 and MISEV2018, respectively. The goal of the current document, MISEV2023, is to provide researchers with an updated snapshot of available approaches and their advantages and limitations for production, separation and characterisation of EVs from multiple sources, including cell culture, body fluids and solid tissues. In addition to presenting the latest state of the art in basic principles of EV research, this document also covers advanced techniques and approaches that are currently expanding the boundaries of the field. MISEV2023 also includes new sections on EV release and uptake and a brief discussion of in vivo approaches to study EVs. Compiling feedback from ISEV expert task forces and more than 1000 researchers, this document conveys the current state of EV research to facilitate robust scientific discoveries and move the field forward even more rapidly.
Collapse
Affiliation(s)
- Joshua A. Welsh
- Translational Nanobiology Section, Laboratory of PathologyNational Cancer Institute, National Institutes of HealthBethesdaMarylandUSA
| | - Deborah C. I. Goberdhan
- Nuffield Department of Women's and Reproductive HealthUniversity of Oxford, Women's Centre, John Radcliffe HospitalOxfordUK
| | - Lorraine O'Driscoll
- School of Pharmacy and Pharmaceutical SciencesTrinity College DublinDublinIreland
- Trinity Biomedical Sciences InstituteTrinity College DublinDublinIreland
- Trinity St. James's Cancer InstituteTrinity College DublinDublinIreland
| | - Edit I. Buzas
- Department of Genetics, Cell‐ and ImmunobiologySemmelweis UniversityBudapestHungary
- HCEMM‐SU Extracellular Vesicle Research GroupSemmelweis UniversityBudapestHungary
- HUN‐REN‐SU Translational Extracellular Vesicle Research GroupSemmelweis UniversityBudapestHungary
| | - Cherie Blenkiron
- Faculty of Medical and Health SciencesThe University of AucklandAucklandNew Zealand
| | - Benedetta Bussolati
- Department of Molecular Biotechnology and Health SciencesUniversity of TurinTurinItaly
| | | | - Dolores Di Vizio
- Department of Surgery, Division of Cancer Biology and TherapeuticsCedars‐Sinai Medical CenterLos AngelesCaliforniaUSA
| | - Tom A. P. Driedonks
- Department CDL ResearchUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Uta Erdbrügger
- University of Virginia Health SystemCharlottesvilleVirginiaUSA
| | - Juan M. Falcon‐Perez
- Exosomes Laboratory, Center for Cooperative Research in BiosciencesBasque Research and Technology AllianceDerioSpain
- Metabolomics Platform, Center for Cooperative Research in BiosciencesBasque Research and Technology AllianceDerioSpain
- IKERBASQUE, Basque Foundation for ScienceBilbaoSpain
| | - Qing‐Ling Fu
- Otorhinolaryngology Hospital, The First Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
- Extracellular Vesicle Research and Clinical Translational CenterThe First Affiliated Hospital, Sun Yat‐sen UniversityGuangzhouChina
| | - Andrew F. Hill
- Institute for Health and SportVictoria UniversityMelbourneAustralia
| | - Metka Lenassi
- Faculty of MedicineUniversity of LjubljanaLjubljanaSlovenia
| | - Sai Kiang Lim
- Institute of Molecular and Cell Biology (IMCB)Agency for Science, Technology and Research (A*STAR)SingaporeSingapore
- Paracrine Therapeutics Pte. Ltd.SingaporeSingapore
- Department of Surgery, YLL School of MedicineNational University SingaporeSingaporeSingapore
| | - Mỹ G. Mahoney
- Thomas Jefferson UniversityPhiladelphiaPennsylvaniaUSA
| | - Sujata Mohanty
- Stem Cell FacilityAll India Institute of Medical SciencesNew DelhiIndia
| | - Andreas Möller
- Chinese University of Hong KongHong KongHong Kong S.A.R.
- QIMR Berghofer Medical Research InstituteBrisbaneAustralia
| | - Rienk Nieuwland
- Laboratory of Experimental Clinical Chemistry, Amsterdam University Medical Centers, Location AMCUniversity of AmsterdamAmsterdamThe Netherlands
- Amsterdam Vesicle Center, Amsterdam University Medical Centers, Location AMCUniversity of AmsterdamAmsterdamThe Netherlands
| | | | - Susmita Sahoo
- Icahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Ana C. Torrecilhas
- Laboratório de Imunologia Celular e Bioquímica de Fungos e Protozoários, Departamento de Ciências Farmacêuticas, Instituto de Ciências Ambientais, Químicas e FarmacêuticasUniversidade Federal de São Paulo (UNIFESP) Campus DiademaDiademaBrazil
| | - Lei Zheng
- Department of Laboratory Medicine, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
| | - Andries Zijlstra
- Department of PathologyVanderbilt University Medical CenterNashvilleTennesseeUSA
- GenentechSouth San FranciscoCaliforniaUSA
| | - Sarah Abuelreich
- Department of Molecular Medicine, Beckman Research InstituteCity of Hope Comprehensive Cancer CenterDuarteCaliforniaUSA
| | - Reem Bagabas
- Department of Molecular Medicine, Beckman Research InstituteCity of Hope Comprehensive Cancer CenterDuarteCaliforniaUSA
| | - Paolo Bergese
- Department of Molecular and Translational MedicineUniversity of BresciaBresciaItaly
- Center for Colloid and Surface Science (CSGI)FlorenceItaly
- National Center for Gene Therapy and Drugs based on RNA TechnologyPaduaItaly
| | - Esther M. Bridges
- Weatherall Institute of Molecular MedicineUniversity of OxfordOxfordUK
| | - Marco Brucale
- Consiglio Nazionale delle Ricerche ‐ Istituto per lo Studio dei Materiali NanostrutturatiBolognaItaly
- Consorzio Interuniversitario per lo Sviluppo dei Sistemi a Grande InterfaseFlorenceItaly
| | - Dylan Burger
- Kidney Research CentreOttawa Hopsital Research InstituteOttawaCanada
- Department of Cellular and Molecular MedicineUniversity of OttawaOttawaCanada
- School of Pharmaceutical SciencesUniversity of OttawaOttawaCanada
| | - Randy P. Carney
- Department of Biomedical EngineeringUniversity of CaliforniaDavisCaliforniaUSA
| | - Emanuele Cocucci
- Division of Pharmaceutics and Pharmacology, College of PharmacyThe Ohio State UniversityColumbusOhioUSA
- Comprehensive Cancer CenterThe Ohio State UniversityColumbusOhioUSA
| | - Federico Colombo
- Division of Pharmaceutics and Pharmacology, College of PharmacyThe Ohio State UniversityColumbusOhioUSA
| | - Rossella Crescitelli
- Sahlgrenska Center for Cancer Research, Department of Surgery, Institute of Clinical SciencesSahlgrenska Academy, University of GothenburgGothenburgSweden
- Wallenberg Centre for Molecular and Translational Medicine, Institute of Clinical SciencesSahlgrenska Academy, University of GothenburgGothenburgSweden
| | - Edveena Hanser
- Department of BiomedicineUniversity Hospital BaselBaselSwitzerland
- Department of BiomedicineUniversity of BaselBaselSwitzerland
| | | | - Norman J. Haughey
- Departments of Neurology and PsychiatryJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - An Hendrix
- Laboratory of Experimental Cancer Research, Department of Human Structure and RepairGhent UniversityGhentBelgium
- Cancer Research Institute GhentGhentBelgium
| | - Alexander R. Ivanov
- Barnett Institute of Chemical and Biological Analysis, Department of Chemistry and Chemical BiologyNortheastern UniversityBostonMassachusettsUSA
| | - Tijana Jovanovic‐Talisman
- Department of Cancer Biology and Molecular Medicine, Beckman Research InstituteCity of Hope Comprehensive Cancer CenterDuarteCaliforniaUSA
| | - Nicole A. Kruh‐Garcia
- Bio‐pharmaceutical Manufacturing and Academic Resource Center (BioMARC)Infectious Disease Research Center, Colorado State UniversityFort CollinsColoradoUSA
| | - Vroniqa Ku'ulei‐Lyn Faustino
- Department of Molecular Medicine, Beckman Research InstituteCity of Hope Comprehensive Cancer CenterDuarteCaliforniaUSA
| | - Diego Kyburz
- Department of BiomedicineUniversity of BaselBaselSwitzerland
- Department of RheumatologyUniversity Hospital BaselBaselSwitzerland
| | - Cecilia Lässer
- Krefting Research Centre, Department of Internal Medicine and Clinical NutritionInstitute of Medicine at Sahlgrenska Academy, University of GothenburgGothenburgSweden
| | - Kathleen M. Lennon
- Department of Molecular Medicine, Beckman Research InstituteCity of Hope Comprehensive Cancer CenterDuarteCaliforniaUSA
| | - Jan Lötvall
- Krefting Research Centre, Institute of Medicine at Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - Adam L. Maddox
- Department of Molecular Medicine, Beckman Research InstituteCity of Hope Comprehensive Cancer CenterDuarteCaliforniaUSA
| | - Elena S. Martens‐Uzunova
- Erasmus MC Cancer InstituteUniversity Medical Center Rotterdam, Department of UrologyRotterdamThe Netherlands
| | - Rachel R. Mizenko
- Department of Biomedical EngineeringUniversity of CaliforniaDavisCaliforniaUSA
| | - Lauren A. Newman
- College of Medicine and Public HealthFlinders UniversityAdelaideAustralia
| | - Andrea Ridolfi
- Department of Physics and Astronomy, and LaserLaB AmsterdamVrije Universiteit AmsterdamAmsterdamThe Netherlands
| | - Eva Rohde
- Department of Transfusion Medicine, University HospitalSalzburger Landeskliniken GmbH of Paracelsus Medical UniversitySalzburgAustria
- GMP Unit, Paracelsus Medical UniversitySalzburgAustria
- Transfer Centre for Extracellular Vesicle Theralytic Technologies, EV‐TTSalzburgAustria
| | - Tatu Rojalin
- Department of Biomedical EngineeringUniversity of CaliforniaDavisCaliforniaUSA
- Expansion Therapeutics, Structural Biology and BiophysicsJupiterFloridaUSA
| | - Andrew Rowland
- College of Medicine and Public HealthFlinders UniversityAdelaideAustralia
| | - Andras Saftics
- Department of Molecular Medicine, Beckman Research InstituteCity of Hope Comprehensive Cancer CenterDuarteCaliforniaUSA
| | - Ursula S. Sandau
- Department of Anesthesiology & Perioperative MedicineOregon Health & Science UniversityPortlandOregonUSA
| | - Julie A. Saugstad
- Department of Anesthesiology & Perioperative MedicineOregon Health & Science UniversityPortlandOregonUSA
| | - Faezeh Shekari
- Department of Stem Cells and Developmental Biology, Cell Science Research CenterRoyan Institute for Stem Cell Biology and Technology, ACECRTehranIran
- Celer DiagnosticsTorontoCanada
| | - Simon Swift
- Waipapa Taumata Rau University of AucklandAucklandNew Zealand
| | - Dmitry Ter‐Ovanesyan
- Wyss Institute for Biologically Inspired EngineeringHarvard UniversityBostonMassachusettsUSA
| | - Juan P. Tosar
- Universidad de la RepúblicaMontevideoUruguay
- Institut Pasteur de MontevideoMontevideoUruguay
| | - Zivile Useckaite
- College of Medicine and Public HealthFlinders UniversityAdelaideAustralia
| | - Francesco Valle
- Consiglio Nazionale delle Ricerche ‐ Istituto per lo Studio dei Materiali NanostrutturatiBolognaItaly
- Consorzio Interuniversitario per lo Sviluppo dei Sistemi a Grande InterfaseFlorenceItaly
| | - Zoltan Varga
- Biological Nanochemistry Research GroupInstitute of Materials and Environmental Chemistry, Research Centre for Natural SciencesBudapestHungary
- Department of Biophysics and Radiation BiologySemmelweis UniversityBudapestHungary
| | - Edwin van der Pol
- Amsterdam Vesicle Center, Amsterdam University Medical Centers, Location AMCUniversity of AmsterdamAmsterdamThe Netherlands
- Biomedical Engineering and Physics, Amsterdam UMC, location AMCUniversity of AmsterdamAmsterdamThe Netherlands
- Laboratory of Experimental Clinical Chemistry, Amsterdam UMC, location AMCUniversity of AmsterdamAmsterdamThe Netherlands
| | - Martijn J. C. van Herwijnen
- Department of Biomolecular Health Sciences, Faculty of Veterinary MedicineUtrecht UniversityUtrechtThe Netherlands
| | - Marca H. M. Wauben
- Department of Biomolecular Health Sciences, Faculty of Veterinary MedicineUtrecht UniversityUtrechtThe Netherlands
| | | | | | - Andrea Zendrini
- Department of Molecular and Translational MedicineUniversity of BresciaBresciaItaly
- Center for Colloid and Surface Science (CSGI)FlorenceItaly
| | - Alan J. Zimmerman
- Barnett Institute of Chemical and Biological Analysis, Department of Chemistry and Chemical BiologyNortheastern UniversityBostonMassachusettsUSA
| | | | - Clotilde Théry
- Institut Curie, INSERM U932PSL UniversityParisFrance
- CurieCoreTech Extracellular Vesicles, Institut CurieParisFrance
| | - Kenneth W. Witwer
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
- EV Core Facility “EXCEL”, Institute for Basic Biomedical SciencesJohns Hopkins University School of MedicineBaltimoreMarylandUSA
- The Richman Family Precision Medicine Center of Excellence in Alzheimer's DiseaseJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| |
Collapse
|
38
|
Malvicini R, De Lazzari G, Tolomeo AM, Santa-Cruz D, Ullah M, Cirillo C, Grumati P, Pacienza N, Muraca M, Yannarelli G. Influence of the isolation method on characteristics and functional activity of mesenchymal stromal cell-derived extracellular vesicles. Cytotherapy 2024; 26:157-170. [PMID: 38069981 DOI: 10.1016/j.jcyt.2023.11.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Revised: 11/07/2023] [Accepted: 11/07/2023] [Indexed: 02/03/2024]
Abstract
BACKGROUND AIMS Extracellular vesicle (EV) isolation methods are based on different physicochemical properties and may result in the purification of distinct EV populations. We compared two different isolation methods suitable for producing clinical-grade mesenchymal stromal cell-derived EVs (MSC-EVs)-ion exchange chromatography (IEX) and ultrafiltration (UF)-and evaluated their impact on the composition and functional properties of EVs. METHODS EVs were purified from conditioned culture medium using an anion exchange resin (IEX) or Amicon filters with a 100-kDa cutoff (UF) (MilliporeSigma, Burlington, MA, USA). We assessed nanoparticle size and distribution by nanoparticle tracking analysis (NTA) and tunable resistive pulse sensing (TRPS) and morphology by transmission electron microscopy. We also measured protein, lipid and total RNA concentration and immunophenotyped both EV populations by flow cytometry (MACSPlex assay; Miltenyi Biotec, Bergisch Gladbach, Germany). Moreover, immunomodulatory activity was tested using a standardized macrophage polarization assay and T-cell stimulation assay. Finally, proteomic analysis and cytokine quantification were carried out to better characterize both EV populations. RESULTS We found by both TRPS and NTA that IEX and UF yielded a comparable amount of total particles with similar size and distribution. In addition, a similar quantity of lipids was obtained with the two procedures. However, IEX yielded 10-fold higher RNA quantity and a larger amount of proteins than UF. MSC-EVs isolated from IEX and UF were positive for the exosome markers CD9, CD63 and CD81 and showed a comparable surface marker expression pattern. Both populations demonstrated immunomodulatory activity in vitro, as they prevented acquisition of the M1 phenotype in lipopolysaccharide-stimulated macrophages and inhibited acquisition of the activation markers CD69 and CD25 on T cells, but the IEX-EVs exerted a significantly greater immunomodulatory effect on both macrophages and T cells compared with UF-EVs. Proteomic analysis and gene ontology enrichment analysis revealed no major differences between the preparations. Finally, cytokine quantification revealed that IEX-EVs were more enriched in some crucial anti-inflammatory and immunomodulatory cytokines (e.g., IL-2, IL-10, transforming growth factor beta and vascular endothelial growth factor) compared with UF-EVs. CONCLUSIONS MSC-EVs isolated by IEX and UF displayed similar physicochemical, phenotypic and functional characteristics. In our conditions, both EV populations demonstrated important anti-inflammatory activity in macrophages and T cells. However, IEX-EVs were more potent than UF-EVs, which may indicate the superiority of this method for the production of clinical-grade EVs.
Collapse
Affiliation(s)
- Ricardo Malvicini
- Laboratorio de Regulación Génica y Células Madre, Instituto de Medicina Traslacional, Trasplante y Bioingeniería, Universidad Favaloro-Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina; Department of Women's and Children's Health, University of Padua, Padua, Italy; Laboratory of Extracellular Vesicles as Therapeutic Tools, Fondazione Istituto di Ricerca Pediatrica Città della Speranza, Padua, Italy; LIFELAB Program, Consorzio per la Ricerca Sanitaria, Padua, Italy.
| | - Giada De Lazzari
- Department of Women's and Children's Health, University of Padua, Padua, Italy; Laboratory of Extracellular Vesicles as Therapeutic Tools, Fondazione Istituto di Ricerca Pediatrica Città della Speranza, Padua, Italy
| | - Anna Maria Tolomeo
- Laboratory of Extracellular Vesicles as Therapeutic Tools, Fondazione Istituto di Ricerca Pediatrica Città della Speranza, Padua, Italy; LIFELAB Program, Consorzio per la Ricerca Sanitaria, Padua, Italy; Department of Cardiac, Thoracic and Vascular Science and Public Health, University of Padova, Padua, Italy
| | - Diego Santa-Cruz
- Laboratorio de Regulación Génica y Células Madre, Instituto de Medicina Traslacional, Trasplante y Bioingeniería, Universidad Favaloro-Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | - Mujib Ullah
- Institute for Immunity and Transplantation, Stem Cell Biology and Regenerative Medicine, School of Medicine, Stanford University, Palo Alto, California, USA
| | - Carmine Cirillo
- Telethon Institute of Genetics and Medicine, Pozzuoli, Naples, Italy
| | - Paolo Grumati
- Telethon Institute of Genetics and Medicine, Pozzuoli, Naples, Italy; Department of Clinical Medicine and Surgery, University of Napoli Federico II, Naples, Italy
| | - Natalia Pacienza
- Laboratorio de Regulación Génica y Células Madre, Instituto de Medicina Traslacional, Trasplante y Bioingeniería, Universidad Favaloro-Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | - Maurizio Muraca
- Department of Women's and Children's Health, University of Padua, Padua, Italy; Laboratory of Extracellular Vesicles as Therapeutic Tools, Fondazione Istituto di Ricerca Pediatrica Città della Speranza, Padua, Italy; LIFELAB Program, Consorzio per la Ricerca Sanitaria, Padua, Italy
| | - Gustavo Yannarelli
- Laboratorio de Regulación Génica y Células Madre, Instituto de Medicina Traslacional, Trasplante y Bioingeniería, Universidad Favaloro-Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina.
| |
Collapse
|
39
|
Qin B, Hu XM, Huang YX, Yang RH, Xiong K. A New Paradigm in Spinal Cord Injury Therapy: from Cell-free Treatment to Engineering Modifications. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2024; 23:656-673. [PMID: 37076458 DOI: 10.2174/1871527322666230418090857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 01/20/2023] [Accepted: 02/06/2023] [Indexed: 04/21/2023]
Abstract
Spinal cord injury (SCI) is an intractable and poorly prognostic neurological disease, and current treatments are still unable to cure it completely and avoid sequelae. Extracellular vesicles (EVs), as important carriers of intercellular communication and pharmacological effects, are considered to be the most promising candidates for SCI therapy because of their low toxicity and immunogenicity, their ability to encapsulate endogenous bioactive molecules (e.g., proteins, lipids, and nucleic acids), and their ability to cross the blood-brain/cerebrospinal barriers. However, poor targeting, low retention rate, and limited therapeutic efficacy of natural EVs have bottlenecked EVs-based SCI therapy. A new paradigm for SCI treatment will be provided by engineering modified EVs. Furthermore, our limited understanding of the role of EVs in SCI pathology hinders the rational design of novel EVbased therapeutic approaches. In this study, we review the pathophysiology after SCI, especially the multicellular EVs-mediated crosstalk; briefly describe the shift from cellular to cell-free therapies for SCI treatment; discuss and analyze the issues related to the route and dose of EVs administration; summarize and present the common strategies for EVs drug loading in the treatment of SCI and point out the shortcomings of these drug loading methods; finally, we analyze and highlight the feasibility and advantages of bio-scaffold-encapsulated EVs for SCI treatment, providing scalable insights into cell-free therapy for SCI.
Collapse
Affiliation(s)
- Bo Qin
- Hubei Key Laboratory for Kidney Disease Pathogenesis and Intervention, Hubei Polytechnic University School of Medicine, Huangshi, 435003, China
| | - Xi-Min Hu
- Clinical Medicine Eight-year Program, 02 Class, 17 Grade, Xiangya School of Medicine, Central South University, Changsha, 410013, China
| | - Yan-Xia Huang
- Health Management Center, the Third Xiangya Hospital, Central South University, Changsha, 410013, China
| | - Rong-Hua Yang
- Department of Burn and Plastic Surgery, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, China
| | - Kun Xiong
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, 410013, China
- Hunan Key Laboratory of Ophthalmology, Changsha, 410008, China
- Key Laboratory of Emergency and Trauma, Ministry of Education, College of Emergency and Trauma, Hainan Medical University, Haikou, 571199, China
| |
Collapse
|
40
|
Petersen AP, Zierden HC. Molecular Approaches to Measure Bacterial Extracellular Vesicles in Tissue Samples. Methods Mol Biol 2024; 2843:253-271. [PMID: 39141305 DOI: 10.1007/978-1-0716-4055-5_16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/15/2024]
Abstract
Extracellular vesicles (EVs) are naturally occurring lipid-bound nanoparticles produced by all cell types. Growing work demonstrates the ability of EVs to facilitate long-distance and cross-kingdom communication. Their innate barrier crossing and cell targeting properties make them a uniquely useful starting ground for novel drug delivery platforms. To better understand the endogenous activity and therapeutic potential of EVs, recent work has measured particle circulation and distribution in vivo using several approaches. Here, we describe molecular-based methods for quantifying bacterial EV distribution in collected tissue samples for biodistribution studies. These methods are important for understanding cell-cell communication facilitated by bacterial EVs and for identifying opportunities for using bacterial EVs as a therapeutic platform.
Collapse
Affiliation(s)
- Alyssa P Petersen
- Department of Chemical & Biomolecular Engineering, University of Maryland, College Park, MD, USA
| | - Hannah C Zierden
- Department of Chemical & Biomolecular Engineering, University of Maryland, College Park, MD, USA.
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, USA.
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
41
|
Shi Y, Wang X, Zhang S, Yin H, Fan H, Tang Y, Yang N. Research progress in in vivo tracing technology for extracellular vesicles. EXTRACELLULAR VESICLES AND CIRCULATING NUCLEIC ACIDS 2023; 4:684-697. [PMID: 39697802 PMCID: PMC11648465 DOI: 10.20517/evcna.2023.49] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 11/20/2023] [Accepted: 12/04/2023] [Indexed: 12/20/2024]
Abstract
Cells have the capability to discharge extracellular vesicles (EVs) into a range of bodily fluids. Extracellular vesicles (EVs) encapsulate biological molecules such as proteins and nucleic acids, playing a role in facilitating cell-cell communication. They actively engage in a myriad of physiological and pathological processes. In vivo tracing of EVs in organisms significantly contributes to elucidating the biological mechanisms of EV-based therapy. The development of molecular imaging technology makes it possible to trace EVs in vivo. Experiments frequently employ a range of molecular imaging techniques, encompassing bioluminescence imaging, fluorescence imaging, magnetic resonance imaging, single photon emission computed tomography, positron emission tomography, photoacoustic imaging, and multimodal imaging. These methods have their own advantages and disadvantages. In this review, typical applications of in vivo tracing of EVs are reviewed.
Collapse
Affiliation(s)
- Yanhua Shi
- School of Bioscience and Technology, Weifang Medical University, Weifang 261053, Shandong, China
- Medical Laboratory Animal Center, Weifang Medical University, Weifang 261053, Shandong, China
- Weifang Key Laboratory of Animal Model Research on Cardiovascular and Cerebrovascular Diseases, Weifang 261053, Shandong, China
- Authors contributed equally
| | - Xianghui Wang
- School of Bioscience and Technology, Weifang Medical University, Weifang 261053, Shandong, China
- School of Biomedical Engineering and Affiliated Sixth People’s Hospital, Shanghai Jiao Tong University, Shanghai 200030, Shanghai, China
- Authors contributed equally
| | - Shifang Zhang
- School of Rehabilitation Medicine, Weifang Medical University, Weifang 261053, Shandong, China
| | - Hao Yin
- School of Bioscience and Technology, Weifang Medical University, Weifang 261053, Shandong, China
| | - Huaju Fan
- School of Psychology, Weifang Medical University, Weifang 261053, Shandong, China
| | - Yaohui Tang
- School of Biomedical Engineering and Affiliated Sixth People’s Hospital, Shanghai Jiao Tong University, Shanghai 200030, Shanghai, China
| | - Nana Yang
- School of Bioscience and Technology, Weifang Medical University, Weifang 261053, Shandong, China
- Medical Laboratory Animal Center, Weifang Medical University, Weifang 261053, Shandong, China
- Weifang Key Laboratory of Animal Model Research on Cardiovascular and Cerebrovascular Diseases, Weifang 261053, Shandong, China
| |
Collapse
|
42
|
Han C, Yang J, Yin T, An J, Qiao A, Cao Y, Feng Y, Lu H, Wang Y, Yang L, Qin G. CD63-Snorkel tagging for isolation of exosomes. EXTRACELLULAR VESICLE 2023; 2:100031. [PMID: 40151378 PMCID: PMC11949438 DOI: 10.1016/j.vesic.2023.100031] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/29/2025]
Abstract
Exosomes (Exo) are important mediators of inter-cellular communications; however, no effective method is available for isolating, thus characterizing, cellular-specific exosomes in vivo. Since CD63 is a reliable marker for exosomes, we have developed a tagging strategy, term "CD63-Snorkel (CD63-SNKL)", in which CD63 at its intracellular C-terminus was fused to a fragment of PDGFRB that contains the transmembrane domain tethered to multiple epitope tags (HA, His, and FLAG) displayed in tandem on surface. We found that the CD63-SNKL protein has similar subcellular localizations as endogenous CD63 and can be effectively sorted into Exo. Furthermore, Exo secreted from CD63-SNKL-transduced cells can be effectively captured on anti-HA magnetic beads and eluted with HA peptides. Thus, CD63-SNKL may be engineered for isolating and tracking endogenous tissue-specific Exo in vivo.
Collapse
Affiliation(s)
- Chaoshan Han
- Department of Pharmacology, Cardiovascular Research Institute, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong 518055, P. R. China
| | - Junjie Yang
- Department of Biomedical Engineering, The University of Alabama at Birmingham, School of Medicine and School of Engineering, Birmingham, AL 35294, USA
| | - Tingting Yin
- Department of Pharmacology, Cardiovascular Research Institute, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong 518055, P. R. China
| | - Junqing An
- Department of Pharmacology, Cardiovascular Research Institute, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong 518055, P. R. China
| | - Aijun Qiao
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, P. R. China
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai 201203, China
| | - Yangpo Cao
- Department of Pharmacology, Cardiovascular Research Institute, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong 518055, P. R. China
| | - Yuliang Feng
- Department of Pharmacology, Cardiovascular Research Institute, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong 518055, P. R. China
| | - Haocheng Lu
- Department of Pharmacology, Cardiovascular Research Institute, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong 518055, P. R. China
| | - Ying Wang
- Department of Pharmacology, Cardiovascular Research Institute, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong 518055, P. R. China
| | - Liang Yang
- School of Medicine, Southern University of Science and Technology, Guangdong 518055, P. R. China
| | - Gangjian Qin
- Department of Pharmacology, Cardiovascular Research Institute, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong 518055, P. R. China
- Department of Biomedical Engineering, The University of Alabama at Birmingham, School of Medicine and School of Engineering, Birmingham, AL 35294, USA
| |
Collapse
|
43
|
Rodriguez BV, Wen Y, Shirk EN, Vazquez S, Gololobova O, Maxwell A, Plunkard J, Castell N, Carlson B, Queen SE, Izzi JM, Driedonks TAP, Witwer KW. An ex vivo model of interactions between extracellular vesicles and peripheral mononuclear blood cells in whole blood. J Extracell Vesicles 2023; 12:e12368. [PMID: 38047476 PMCID: PMC10694845 DOI: 10.1002/jev2.12368] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 08/25/2023] [Accepted: 09/13/2023] [Indexed: 12/05/2023] Open
Abstract
Extracellular vesicles (EVs) can be loaded with therapeutic cargo and engineered for retention by specific body sites; therefore, they have great potential for targeted delivery of biomolecules to treat diseases. However, the pharmacokinetics and biodistribution of EVs in large animals remain relatively unknown, especially in primates. We recently reported that when cell culture-derived EVs are administered intravenously to Macaca nemestrina (pig-tailed macaques), they differentially associate with specific subsets of peripheral blood mononuclear cells (PBMCs). More than 60% of CD20+ B cells were observed to associate with EVs for up to 1 h post-intravenous administration. To investigate these associations further, we developed an ex vivo model of whole blood collected from healthy pig-tailed macaques. Using this ex vivo system, we found that labelled EVs preferentially associate with B cells in whole blood at levels similar to those detected in vivo. This study demonstrates that ex vivo blood can be used to study EV-blood cell interactions.
Collapse
Affiliation(s)
- Blanca V. Rodriguez
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Yi Wen
- 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
| | - Samuel Vazquez
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Olesia Gololobova
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Amanda Maxwell
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Jessica Plunkard
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Natalie Castell
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Bess Carlson
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Suzanne E. Queen
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Jessica M. Izzi
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Tom A. P. Driedonks
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
- University Medical CenterUtrecht UniversityUtrechtThe Netherlands
| | - Kenneth W. Witwer
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
- Department of NeurologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
- Richman Family Precision Medicine Center of Excellence in Alzheimer's DiseaseJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| |
Collapse
|
44
|
Koksal AR, Ekmen N, Aydin Y, Nunez K, Sandow T, Delk M, Moehlen M, Thevenot P, Cohen A, Dash S. A Single-Step Immunocapture Assay to Quantify HCC Exosomes Using the Highly Sensitive Fluorescence Nanoparticle-Tracking Analysis. J Hepatocell Carcinoma 2023; 10:1935-1954. [PMID: 37936599 PMCID: PMC10627088 DOI: 10.2147/jhc.s423043] [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: 06/15/2023] [Accepted: 10/07/2023] [Indexed: 11/09/2023] Open
Abstract
Introduction Extracellular vesicles could serve as a non-invasive biomarker for early cancer detection. However, limited methods to quantitate cancer-derived vesicles in the native state remain a significant barrier to clinical translation. Aim This research aims to develop a rapid, one-step immunoaffinity approach to quantify HCC exosomes directly from a small serum volume. Methods HCC-derived exosomes in the serum were captured using fluorescent phycoerythrin (PE)-conjugated antibodies targeted to GPC3 and alpha-fetoprotein (AFP). Total and HCC-specific exosomes were then quantified in culture supernatant or patient-derived serums using fluorescence nanoparticle tracking analysis (F-NTA). The performance of HCC exosome quantification in the serum was compared with the tumor size determined by MRI. Results Initially we tested the detection limits of the F-NTA using synthetic fluorescent and non-fluorescent beads. The assay showed an acceptable sensitivity with a detection range of 104-108 particles/mL. Additionally, the combination of immunocapture followed by size-exclusion column purification allows the isolation of smaller-size EVs and quantification by F-NTA. Our assay demonstrated that HCC cell culture releases a significantly higher quantity of GPC3 or GPC3+AFP positive EVs (100-200 particles/cell) compared to non-HCC culture (10-40 particles/cell) (p<0.01 and p<0.05 respectively). The F-NTA enables absolute counting of HCC-specific exosomes in the clinical samples with preserved biological immunoreactivity. The performance of F-NTA was clinically validated in serum from patients ± cirrhosis and with confirmed HCC. F-NTA quantification data show selective enrichment of AFP and GPC3 positive EVs in HCC serum compared to malignancy-free cirrhosis (AUC values for GPC3, AFP, and GPC3/AFP were found 0.79, 0.71, and 0.72 respectively). The MRI-confirmed patient cohort indicated that there was a positive correlation between total tumor size and GPC3-positive exosome concentration (r:0.78 and p<0.001). Conclusion We developed an immunocapture assay that can be used for simultaneous isolation and quantification of HCC-derived exosomes from a small serum volume with high accuracy.
Collapse
Affiliation(s)
- Ali Riza Koksal
- Department of Pathology and Laboratory Medicine, Tulane University Health Sciences Center, New Orleans, LA, USA
| | - Nergiz Ekmen
- Department of Gastroenterology and Hepatology, Tulane University Health Sciences Center, New Orleans, LA, USA
| | - Yucel Aydin
- Department of Gastroenterology and Hepatology, Tulane University Health Sciences Center, New Orleans, LA, USA
| | - Kelley Nunez
- Department of Gastroenterology and Hepatology, Institute of Translational Research, Ochsner Health, New Orleans, LA, USA
| | - Tyler Sandow
- Department of Radiology, Institute of Translational Research, Ochsner Health, New Orleans, LA, USA
| | - Molly Delk
- Department of Gastroenterology and Hepatology, Tulane University Health Sciences Center, New Orleans, LA, USA
| | - Martin Moehlen
- Department of Gastroenterology and Hepatology, Tulane University Health Sciences Center, New Orleans, LA, USA
| | - Paul Thevenot
- Department of Gastroenterology and Hepatology, Institute of Translational Research, Ochsner Health, New Orleans, LA, USA
| | - Ari Cohen
- Department of Gastroenterology and Hepatology, Institute of Translational Research, Ochsner Health, New Orleans, LA, USA
- Multi-Organ Transplant Institute, Ochsner Health, New Orleans, LA, USA
| | - Srikanta Dash
- Department of Pathology and Laboratory Medicine, Tulane University Health Sciences Center, New Orleans, LA, USA
- Department of Gastroenterology and Hepatology, Tulane University Health Sciences Center, New Orleans, LA, USA
- Southeast Louisiana Veterans Health Care System, New Orleans, LA, USA
| |
Collapse
|
45
|
Lucchetti D, Colella F, Artemi G, Haque S, Sgambato A, Pellicano R, Fagoonee S. Smart nano-sized extracellular vesicles for cancer therapy: Potential theranostic applications in gastrointestinal tumors. Crit Rev Oncol Hematol 2023; 191:104121. [PMID: 37690633 DOI: 10.1016/j.critrevonc.2023.104121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 01/27/2023] [Accepted: 09/04/2023] [Indexed: 09/12/2023] Open
Abstract
Extracellular vesicles (EVs) have gained tremendous interest in the search for next-generation therapeutics for the treatment of a range of pathologies, including cancer, especially due to their small size, biomolecular cargo, ability to mediate intercellular communication, high physicochemical stability, low immunogenicity and biocompatibility. The theranostic potential of EVs have been enhanced by adopting several strategies such as genetic or metabolic engineering, parental cell modification or direct functionalization to incorporate therapeutic compounds into these nanoplatforms. The smart nano-sized EVs indeed offer huge opportunities in the field of cancer, and current research is set at overcoming the existing pitfalls. Smart EVs are already being applied in the clinics despite the challenges faced. We provide, herein, an update on the technologies employed for EV functionalization in order to achieve optimal tumor cell targeting and EV tracking in vivo with bio-imaging modalities, as well as the preclinical and clinical studies making use of these modified EVs, in the context of gastrointestinal tumors.
Collapse
Affiliation(s)
- Donatella Lucchetti
- Fondazione Policlinico Universitario 'Agostino Gemelli' IRCCS, Rome, Italy; Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Filomena Colella
- Fondazione Policlinico Universitario 'Agostino Gemelli' IRCCS, Rome, Italy
| | - Giulia Artemi
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Shafiul Haque
- Research and Scientific Studies Unit, College of Nursing and Allied Health Sciences, Jazan University, Jazan 45142, Saudi Arabia; Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Beirut 1102 2801, Lebanon; Centre of Medical and Bio-Allied Health Sciences Research, Ajman University, Ajman 13306, United Arab Emirates
| | - Alessandro Sgambato
- Fondazione Policlinico Universitario 'Agostino Gemelli' IRCCS, Rome, Italy; Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy.
| | - Rinaldo Pellicano
- Gastroenterology Unit, Città della salute e della Scienza Hospital, Turin, Italy
| | - Sharmila Fagoonee
- Institute of Biostructure and Bioimaging (CNR), Molecular Biotechnology Center, Turin, Italy
| |
Collapse
|
46
|
Amini H, Namjoo AR, Narmi MT, Mardi N, Narimani S, Naturi O, Khosrowshahi ND, Rahbarghazi R, Saghebasl S, Hashemzadeh S, Nouri M. Exosome-bearing hydrogels and cardiac tissue regeneration. Biomater Res 2023; 27:99. [PMID: 37803483 PMCID: PMC10559618 DOI: 10.1186/s40824-023-00433-3] [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: 05/26/2023] [Accepted: 09/18/2023] [Indexed: 10/08/2023] Open
Abstract
BACKGROUND In recent years, cardiovascular disease in particular myocardial infarction (MI) has become the predominant cause of human disability and mortality in the clinical setting. The restricted capacity of adult cardiomyocytes to proliferate and restore the function of infarcted sites is a challenging issue after the occurrence of MI. The application of stem cells and byproducts such as exosomes (Exos) has paved the way for the alleviation of cardiac tissue injury along with conventional medications in clinics. However, the short lifespan and activation of alloreactive immune cells in response to Exos and stem cells are the main issues in patients with MI. Therefore, there is an urgent demand to develop therapeutic approaches with minimum invasion for the restoration of cardiac function. MAIN BODY Here, we focused on recent data associated with the application of Exo-loaded hydrogels in ischemic cardiac tissue. Whether and how the advances in tissue engineering modalities have increased the efficiency of whole-based and byproducts (Exos) therapies under ischemic conditions. The integration of nanotechnology and nanobiology for designing novel smart biomaterials with therapeutic outcomes was highlighted. CONCLUSION Hydrogels can provide suitable platforms for the transfer of Exos, small molecules, drugs, and other bioactive factors for direct injection into the damaged myocardium. Future studies should focus on the improvement of physicochemical properties of Exo-bearing hydrogel to translate for the standard treatment options.
Collapse
Affiliation(s)
- Hassan Amini
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of General and Vascular Surgery, Tabriz University of Medical Sciences, Tabriz, 51548/53431, Iran
| | - Atieh Rezaei Namjoo
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Maryam Taghavi Narmi
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Narges Mardi
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Samaneh Narimani
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ozra Naturi
- Department of Organic and Biochemistry, Faculty of Chemistry, University of Tabriz, Tabriz, Iran
| | - Nafiseh Didar Khosrowshahi
- Stem Cell and Tissue Engineering Research Laboratory, Sahand University of Technology, Tabriz, 51335-1996, Iran
| | - Reza Rahbarghazi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
- Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, 51548/53431, Iran.
| | - Solmaz Saghebasl
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
- Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, 51548/53431, Iran.
| | - Shahriar Hashemzadeh
- Department of General and Vascular Surgery, Tabriz University of Medical Sciences, Tabriz, 51548/53431, Iran.
| | - Mohammad Nouri
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
47
|
Cheng Z, Shang J, Wang H, Yu L, Yuan Z, Zhang Y, Du Y, Tian J. Molecular imaging-guided extracellular vesicle-based drug delivery for precise cancer management: Current status and future perspectives. J Control Release 2023; 362:97-120. [PMID: 37625599 DOI: 10.1016/j.jconrel.2023.08.040] [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: 05/18/2023] [Revised: 08/16/2023] [Accepted: 08/20/2023] [Indexed: 08/27/2023]
Abstract
Extracellular vesicles (EVs), the mediators of intercellular communication, have attracted the attention of researchers for the important roles they play in cancer treatment. Compared with other inorganic nano-materials, EVs possess the advantages of higher biocompatibility, better physiochemical stability, easier surface modification, and excellent biosafety. They can be used as an advanced drug delivery system with an improved therapeutic index for various therapeutic agents. Engineered EV-based imaging and therapeutic agents (engineered EVs) have emerged as useful tools in targeted cancer diagnosis and therapy. Non-invasive tracing of engineered EVs contributes to a better evaluation of their functions in cancer progression, in vivo dynamic biodistribution, therapeutic response, and drug-loading efficiency. Recent advances in real-time molecular imaging (MI), and innovative EV labeling strategies have led to the development of novel tools that can evaluate the pharmacokinetics of engineered EVs in cancer management, which may accelerate further clinical translation of novel EV-based drug delivery platforms. Herein, we review the latest advances in EVs, their characteristics, and current examples of EV-based targeted drug delivery for cancer. Then, we discuss the prominent applications of MI for tracing both natural and engineered EVs. Finally, we discuss the current challenges and considerations of EVs in targeted cancer treatment and the limitations of different MI modalities. In the coming decades, EV-based therapeutic applications for cancer with improved drug loading and targeting abilities will be developed, and better anti-cancer effects of drug delivery nanoplatform will be achieved.
Collapse
Affiliation(s)
- Zhongquan Cheng
- Department of General Surgery, Capital Medical University, Beijing Friendship Hospital, Beijing 100050, China; CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China
| | - Jihuan Shang
- School of Clinical Medicine, Nanjing Medical University, Nanjing 211166, China
| | - Huarong Wang
- Department of General Surgery, Capital Medical University, Beijing Friendship Hospital, Beijing 100050, China
| | - Leyi Yu
- Beijing Haidian Hospital, Beijing 100080, China
| | - Zhu Yuan
- Department of General Surgery, Capital Medical University, Beijing Friendship Hospital, Beijing 100050, China.
| | - Yinlong Zhang
- School of Nanoscience and Engineering, University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Yang Du
- CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China; University of Chinese Academy of Sciences, Beijing 100080, China.
| | - Jie Tian
- CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China; Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, School of Medicine, China; Science and Engineering, Beihang University, Beijing 100191, China.
| |
Collapse
|
48
|
Anastasi F, Botto A, Immordino B, Giovannetti E, McDonnell LA. Proteomics analysis of circulating small extracellular vesicles: Focus on the contribution of EVs to tumor metabolism. Cytokine Growth Factor Rev 2023; 73:3-19. [PMID: 37652834 DOI: 10.1016/j.cytogfr.2023.08.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 08/16/2023] [Indexed: 09/02/2023]
Abstract
The term small extracellular vesicle (sEV) is a comprehensive term that includes any type of cell-derived, membrane-delimited particle that has a diameter < 200 nm, and which includes exosomes and smaller microvesicles. sEVs transfer bioactive molecules between cells and are crucial for cellular homeostasis and particularly during tumor development, where sEVs provide important contributions to the formation of the premetastic niche and to their altered metabolism. sEVs are thus legitimate targets for intervention and have also gained increasing interest as an easily accessible source of biomarkers because they can be rapidly isolated from serum/plasma and their molecular cargo provides information on their cell-of origin. To target sEVs that are specific for a given cell/disease it is essential to identify EV surface proteins that are characteristic of that cell/disease. Mass-spectrometry based proteomics is widely used for the identification and quantification of sEV proteins. The methods used for isolating the sEVs, preparing the sEV sample for proteomics analysis, and mass spectrometry analysis, can have a strong influence on the results and requires careful consideration. This review provides an overview of the approaches used for sEV proteomics and discusses the inherent compromises regarding EV purity versus depth of coverage. Additionally, it discusses the practical applications of the methods to unravel the involvement of sEVs in regulating the metabolism of pancreatic ductal adenocarcinoma (PDAC). The metabolic reprogramming in PDAC includes enhanced glycolysis, elevated glutamine metabolism, alterations in lipid metabolism, mitochondrial dysfunction and hypoxia, all of which are crucial in promoting tumor cell growth. A thorough understanding of these metabolic adaptations is imperative for the development of targeted therapies to exploit PDAC's vulnerabilities.
Collapse
Affiliation(s)
- Federica Anastasi
- Fondazione Pisana per la Scienza ONLUS, San Giuliano Terme, PI, Italy; National Enterprise for NanoScience and NanoTechnology, Scuola Normale Superiore, Pisa, Italy; BarcelonaBeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain
| | - Asia Botto
- Fondazione Pisana per la Scienza ONLUS, San Giuliano Terme, PI, Italy; Department of Chemistry and Industrial Chemistry, University of Pisa, Pisa, Italy
| | - Benoit Immordino
- Fondazione Pisana per la Scienza ONLUS, San Giuliano Terme, PI, Italy; Scuola Superiore Sant'Anna, Pisa, Italy
| | - Elisa Giovannetti
- Fondazione Pisana per la Scienza ONLUS, San Giuliano Terme, PI, Italy; Department of Medical Oncology, Amsterdam UMC, Cancer Center Amsterdam, Vrije Universiteit, Amsterdam, the Netherlands
| | - Liam A McDonnell
- Fondazione Pisana per la Scienza ONLUS, San Giuliano Terme, PI, Italy.
| |
Collapse
|
49
|
Liu B, Nguyen PL, Yu H, Li X, Wang H, Price J, Niu M, Guda C, Cheng X, Sun X, Moreau R, Ramer-Tait A, Naldrett MJ, Alvarez S, Yu J. Critical contributions of protein cargos to the functions of macrophage-derived extracellular vesicles. J Nanobiotechnology 2023; 21:352. [PMID: 37770932 PMCID: PMC10537535 DOI: 10.1186/s12951-023-02105-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 09/13/2023] [Indexed: 09/30/2023] Open
Abstract
BACKGROUND Macrophages are highly plastic innate immune cells that play key roles in host defense, tissue repair, and homeostasis maintenance. In response to divergent stimuli, macrophages rapidly alter their functions and manifest a wide polarization spectrum with two extremes: M1 or classical activation and M2 or alternative activation. Extracellular vesicles (EVs) secreted from differentially activated macrophages have been shown to have diverse functions, which are primarily attributed to their microRNA cargos. The role of protein cargos in these EVs remains largely unexplored. Therefore, in this study, we focused on the protein cargos in macrophage-derived EVs. RESULTS Naïve murine bone marrow-derived macrophages were treated with lipopolysaccharide or interlukin-4 to induce M1 or M2 macrophages, respectively. The proteins of EVs and their parental macrophages were subjected to quantitative proteomics analyses, followed by bioinformatic analyses. The enriched proteins of M1-EVs were involved in proinflammatory pathways and those of M2-EVs were associated with immunomodulation and tissue remodeling. The signature proteins of EVs shared a limited subset of the proteins of their respective progenitor macrophages, but they covered many of the typical pathways and functions of their parental cells, suggesting their respective M1-like and M2-like phenotypes and functions. Experimental examination validated that protein cargos in M1- or M2-EVs induced M1 or M2 polarization, respectively. More importantly, proteins in M1-EVs promoted viability, proliferation, and activation of T lymphocytes, whereas proteins in M2-EVs potently protected the tight junction structure and barrier integrity of epithelial cells from disruption. Intravenous administration of M2-EVs in colitis mice led to their accumulation in the colon, alleviation of colonic inflammation, promotion of M2 macrophage polarization, and improvement of gut barrier functions. Protein cargos in M2-EVs played a key role in their protective function in colitis. CONCLUSION This study has yielded a comprehensive unbiased dataset of protein cargos in macrophage-derived EVs, provided a systemic view of their potential functions, and highlighted the important engagement of protein cargos in the pathophysiological functions of these EVs.
Collapse
Affiliation(s)
- Baolong Liu
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, 230 Filley Hall, Lincoln, NE, 68583, USA
| | - Phuong Linh Nguyen
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, 230 Filley Hall, Lincoln, NE, 68583, USA
| | - Han Yu
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, 230 Filley Hall, Lincoln, NE, 68583, USA
| | - Xingzhi Li
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, 230 Filley Hall, Lincoln, NE, 68583, USA
| | - Huiren Wang
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, 230 Filley Hall, Lincoln, NE, 68583, USA
| | - Jeffrey Price
- Department of Food Science and Technology, University of Nebraska-Lincoln, 260 Food Innovation Center, Lincoln, NE, 68588, USA
- Nebraska Food for Health Center, University of Nebraska-Lincoln, 115 Food Innovation Center, Lincoln, NE, 68588, USA
| | - Meng Niu
- Department of Genetics, Cell Biology and Anatomy, Bioinformatics and Systems Biology Core, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Chittibabu Guda
- Department of Genetics, Cell Biology and Anatomy, Bioinformatics and Systems Biology Core, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Xiao Cheng
- Department of Biochemistry, University of Nebraska-Lincoln, N158 Beadle Center, Lincoln, NE, 68588-0665, USA
| | - Xinghui Sun
- Department of Biochemistry, University of Nebraska-Lincoln, N158 Beadle Center, Lincoln, NE, 68588-0665, USA
| | - Regis Moreau
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, 316E Ruth Leverton Hall, Lincoln, NE, 68583, USA
| | - Amanda Ramer-Tait
- Department of Food Science and Technology, University of Nebraska-Lincoln, 260 Food Innovation Center, Lincoln, NE, 68588, USA
- Nebraska Food for Health Center, University of Nebraska-Lincoln, 115 Food Innovation Center, Lincoln, NE, 68588, USA
| | - Michael J Naldrett
- Proteomics and Metabolomics Facility, Nebraska Center for Biotechnology, University of Nebraska-Lincoln, Lincoln, NE, 68588, USA
| | - Sophie Alvarez
- Proteomics and Metabolomics Facility, Nebraska Center for Biotechnology, University of Nebraska-Lincoln, Lincoln, NE, 68588, USA
| | - Jiujiu Yu
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, 230 Filley Hall, Lincoln, NE, 68583, USA.
| |
Collapse
|
50
|
Ilahibaks NF, Roefs MT, Brans MAD, Blok CS, de Jager SCA, Schiffelers RM, Vader P, Lei Z, Sluijter JPG. Extracellular vesicle-mediated protein delivery to the liver. JOURNAL OF EXTRACELLULAR BIOLOGY 2023; 2:e97. [PMID: 38938374 PMCID: PMC11080727 DOI: 10.1002/jex2.97] [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: 01/16/2023] [Revised: 05/27/2023] [Accepted: 06/08/2023] [Indexed: 06/29/2024]
Abstract
Extracellular vesicles (EVs) are nanoscale particles that facilitate intercellular communication. They are regarded as a promising natural drug delivery system for transporting and delivering bioactive macromolecules to target cells. Recently, researchers have engineered EVs with FKBP12/FRB heterodimerization domains that interact with rapamycin to load and deliver exogenous proteins for both in vitro and in vivo applications. In this study, we examined the tissue distribution of EVs using near-infrared fluorescent imaging. We evaluated the effectiveness of EV-mediated delivery of Cre recombinase specifically to hepatocytes in the livers of Ai9 Cre-loxP reporter mice. Intravenous injection resulted in more efficient Cre protein delivery to the liver than intraperitoneal injections. Depleting liver-resident macrophages with clodronate-encapsulated liposome pre-treatment did not enhance EV-mediated Cre delivery to hepatocytes. Moreover, we demonstrated that multiple intravenous injections of Cre-EVs facilitated functional Cre delivery to hepatocytes. To the best of our knowledge, this is the first study to simultaneously investigate the tissue distribution of FKBP12/FRB-engineered EVs and their subsequent intracellular protein delivery in Ai9 Cre-loxP reporter mice. These insights can inform preclinical research and contribute to developing next-generation EV-based platforms for delivering therapeutic proteins or genome editing technologies targeting the liver.
Collapse
Affiliation(s)
- Nazma F. Ilahibaks
- Laboratory of Experimental Cardiology, Department Heart & LungsUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Marieke T. Roefs
- Laboratory of Experimental Cardiology, Department Heart & LungsUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Maike A. D. Brans
- Laboratory of Experimental Cardiology, Department Heart & LungsUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Christian Snijders Blok
- Laboratory of Experimental Cardiology, Department Heart & LungsUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Saskia C. A. de Jager
- Laboratory of Experimental Cardiology, Department Heart & LungsUniversity Medical Center UtrechtUtrechtThe Netherlands
| | | | - Pieter Vader
- Laboratory of Experimental Cardiology, Department Heart & LungsUniversity Medical Center UtrechtUtrechtThe Netherlands
- CDL ResearchUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Zhiyong Lei
- Laboratory of Experimental Cardiology, Department Heart & LungsUniversity Medical Center UtrechtUtrechtThe Netherlands
- CDL ResearchUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Joost P. G. Sluijter
- Laboratory of Experimental Cardiology, Department Heart & LungsUniversity Medical Center UtrechtUtrechtThe Netherlands
- Circulatory Health Laboratory, Regenerative Medicine CenterUniversity Medical Center Utrecht, University UtrechtUtrechtThe Netherlands
| |
Collapse
|