1051
|
Fuentes-Rodriguez A, Mitchell A, Guérin SL, Landreville S. Recent Advances in Molecular and Genetic Research on Uveal Melanoma. Cells 2024; 13:1023. [PMID: 38920653 PMCID: PMC11201764 DOI: 10.3390/cells13121023] [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: 06/08/2024] [Accepted: 06/09/2024] [Indexed: 06/27/2024] Open
Abstract
Uveal melanoma (UM), a distinct subtype of melanoma, presents unique challenges in its clinical management due to its complex molecular landscape and tendency for liver metastasis. This review highlights recent advancements in understanding the molecular pathogenesis, genetic alterations, and immune microenvironment of UM, with a focus on pivotal genes, such as GNAQ/11, BAP1, and CYSLTR2, and delves into the distinctive genetic and chromosomal classifications of UM, emphasizing the role of mutations and chromosomal rearrangements in disease progression and metastatic risk. Novel diagnostic biomarkers, including circulating tumor cells, DNA and extracellular vesicles, are discussed, offering potential non-invasive approaches for early detection and monitoring. It also explores emerging prognostic markers and their implications for patient stratification and personalized treatment strategies. Therapeutic approaches, including histone deacetylase inhibitors, MAPK pathway inhibitors, and emerging trends and concepts like CAR T-cell therapy, are evaluated for their efficacy in UM treatment. This review identifies challenges in UM research, such as the limited treatment options for metastatic UM and the need for improved prognostic tools, and suggests future directions, including the discovery of novel therapeutic targets, immunotherapeutic strategies, and advanced drug delivery systems. The review concludes by emphasizing the importance of continued research and innovation in addressing the unique challenges of UM to improve patient outcomes and develop more effective treatment strategies.
Collapse
Affiliation(s)
- Aurélie Fuentes-Rodriguez
- Department of Ophthalmology and Otorhinolaryngology-Cervico-Facial Surgery, Faculty of Medicine, Université Laval, Quebec City, QC G1V 0A6, Canada; (A.F.-R.); (A.M.); (S.L.G.)
- Hôpital du Saint-Sacrement, Regenerative Medicine Division, CHU de Québec-Université Laval Research Centre, Quebec City, QC G1S 4L8, Canada
- Centre de Recherche en Organogénèse Expérimentale de l‘Université Laval/LOEX, Quebec City, QC G1J 1Z4, Canada
- Université Laval Cancer Research Center, Quebec City, QC G1R 3S3, Canada
| | - Andrew Mitchell
- Department of Ophthalmology and Otorhinolaryngology-Cervico-Facial Surgery, Faculty of Medicine, Université Laval, Quebec City, QC G1V 0A6, Canada; (A.F.-R.); (A.M.); (S.L.G.)
- Hôpital du Saint-Sacrement, Regenerative Medicine Division, CHU de Québec-Université Laval Research Centre, Quebec City, QC G1S 4L8, Canada
- Centre de Recherche en Organogénèse Expérimentale de l‘Université Laval/LOEX, Quebec City, QC G1J 1Z4, Canada
- Université Laval Cancer Research Center, Quebec City, QC G1R 3S3, Canada
| | - Sylvain L. Guérin
- Department of Ophthalmology and Otorhinolaryngology-Cervico-Facial Surgery, Faculty of Medicine, Université Laval, Quebec City, QC G1V 0A6, Canada; (A.F.-R.); (A.M.); (S.L.G.)
- Hôpital du Saint-Sacrement, Regenerative Medicine Division, CHU de Québec-Université Laval Research Centre, Quebec City, QC G1S 4L8, Canada
- Centre de Recherche en Organogénèse Expérimentale de l‘Université Laval/LOEX, Quebec City, QC G1J 1Z4, Canada
| | - Solange Landreville
- Department of Ophthalmology and Otorhinolaryngology-Cervico-Facial Surgery, Faculty of Medicine, Université Laval, Quebec City, QC G1V 0A6, Canada; (A.F.-R.); (A.M.); (S.L.G.)
- Hôpital du Saint-Sacrement, Regenerative Medicine Division, CHU de Québec-Université Laval Research Centre, Quebec City, QC G1S 4L8, Canada
- Centre de Recherche en Organogénèse Expérimentale de l‘Université Laval/LOEX, Quebec City, QC G1J 1Z4, Canada
- Université Laval Cancer Research Center, Quebec City, QC G1R 3S3, Canada
| |
Collapse
|
1052
|
Zhai C, Xu J, Yang Y, Xie F, Cao L, Wang K, Zhou Y, Ding X, Yin J, Ding X, Hu H, Yu H. Heterogeneous Analysis of Extracellular Vesicles for Osteosarcoma Diagnosis. Anal Chem 2024; 96:9486-9492. [PMID: 38814722 DOI: 10.1021/acs.analchem.4c00941] [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: 06/01/2024]
Abstract
Osteosarcoma (OS) is the most prevalent primary tumor of bones, often diagnosed late with a poor prognosis. Currently, few effective biomarkers or diagnostic methods have been developed for early OS detection with high confidence, especially for metastatic OS. Tumor-derived extracellular vesicles (EVs) are emerging as promising biomarkers for early cancer diagnosis through liquid biopsy. Here, we report a plasmonic imaging-based biosensing technique, termed subpopulation protein analysis by single EV counting (SPASEC), for size-dependent EV subpopulation analysis. In our SPASEC platform, EVs are accurately sized and counted on plasmonic sensor chips coated with OS-specific antibodies. Subsequently, EVs are categorized into distinct subpopulations based on their sizes, and the membrane proteins of each size-dependent subpopulation are profiled. We measured the heterogeneous expression levels of the EV markers (CD63, BMP2, GD2, and N-cadherin) in each of the EV subsets from both OS cell lines and clinical plasma samples. Using the linear discriminant analysis (LDA) model, the combination of four markers is applied to classify the healthy donors (n = 37), nonmetastatic OS patients (n = 13), and metastatic patients (n = 12) with an area under the curve of 0.95, 0.92, and 0.99, respectively. SPASEC provides accurate EV sensing technology for early OS diagnosis.
Collapse
Affiliation(s)
- Chunhui Zhai
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Jiaying Xu
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Yuting Yang
- School of Sensing Science and Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Feng Xie
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Li Cao
- Shanghai Clinical Research Ward (SCRW), Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Kai Wang
- Shanghai Clinical Research Ward (SCRW), Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Yan Zhou
- Department of Oncology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Xiaomin Ding
- Shanghai Clinical Research Ward (SCRW), Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Junyi Yin
- Oncology Department of Tongji Hospital of Tongji University, No. 389 Xincun Road, Shanghai, 200065, China
| | - Xianting Ding
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Haiyan Hu
- Shanghai Clinical Research Ward (SCRW), Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Hui Yu
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| |
Collapse
|
1053
|
Petronio Petronio G, Di Naro M, Venditti N, Guarnieri A, Cutuli MA, Magnifico I, Medoro A, Foderà E, Passarella D, Nicolosi D, Di Marco R. Targeting S. aureus Extracellular Vesicles: A New Putative Strategy to Counteract Their Pathogenic Potential. Pharmaceutics 2024; 16:789. [PMID: 38931910 PMCID: PMC11207539 DOI: 10.3390/pharmaceutics16060789] [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: 04/24/2024] [Revised: 05/27/2024] [Accepted: 06/07/2024] [Indexed: 06/28/2024] Open
Abstract
Long-term inflammatory skin disease atopic dermatitis is characterized by dry skin, itching, and eczematous lesions. During inflammation skin barrier protein impairment promotes S. aureus colonisation in the inflamed skin, worsening AD patient's clinical condition. Proteomic analysis revealed the presence of several immune evasion proteins and virulence factors in S. aureus extracellular vesicles (EVs), suggesting a possible role for these proteins in the pathophysiology of atopic dermatitis. The objective of this study is to assess the efficacy of a wall fragment obtained from a patented strain of C. acnes DSM28251 (c40) and its combination with a mucopolysaccharide carrier (HAc40) in counteract the pathogenic potential of EVs produced by S. aureus ATCC 14458. Results obtained from in vitro studies on HaCaT keratinocyte cells showed that HAc40 and c40 treatment significantly altered the size and pathogenicity of S. aureus EVs. Specifically, EVs grew larger, potentially reducing their ability to interact with the target cells and decreasing cytotoxicity. Additionally, the overexpression of the tight junctions mRNA zona occludens 1 (ZO1) and claudin 1 (CLDN1) following EVs exposure was decreased by HAc40 and c40 treatment, indicating a protective effect on the epidermal barrier's function. These findings demonstrate how Hac40 and c40 may mitigate the harmful effects of S. aureus EVs. Further investigation is needed to elucidate the exact mechanisms underlying this interaction and explore the potential clinical utility of c40 and its mucopolysaccharide carrier conjugate HAc40 in managing atopic dermatitis.
Collapse
Affiliation(s)
- Giulio Petronio Petronio
- Department of Medicina e Scienze della Salute “V. Tiberio”, Università degli Studi del Molise, 86100 Campobasso, Italy (R.D.M.)
| | - Maria Di Naro
- Department of Drug and Health Sciences, Università degli Studi di Catania, 95125 Catania, Italy
| | - Noemi Venditti
- Department of Medicina e Scienze della Salute “V. Tiberio”, Università degli Studi del Molise, 86100 Campobasso, Italy (R.D.M.)
- UO Laboratorio Analisi, Responsible Research Hospital, 86100 Campobasso, Italy
| | - Antonio Guarnieri
- Department of Medicina e Scienze della Salute “V. Tiberio”, Università degli Studi del Molise, 86100 Campobasso, Italy (R.D.M.)
| | | | | | - Alessandro Medoro
- Department of Medicina e Scienze della Salute “V. Tiberio”, Università degli Studi del Molise, 86100 Campobasso, Italy (R.D.M.)
| | - Emanuele Foderà
- Department of Medicina e Scienze della Salute “V. Tiberio”, Università degli Studi del Molise, 86100 Campobasso, Italy (R.D.M.)
| | - Daniela Passarella
- Department of Medicina e Scienze della Salute “V. Tiberio”, Università degli Studi del Molise, 86100 Campobasso, Italy (R.D.M.)
| | - Daria Nicolosi
- Department of Drug and Health Sciences, Università degli Studi di Catania, 95125 Catania, Italy
| | - Roberto Di Marco
- Department of Medicina e Scienze della Salute “V. Tiberio”, Università degli Studi del Molise, 86100 Campobasso, Italy (R.D.M.)
| |
Collapse
|
1054
|
White KE, Bailey HL, Shaw BS, Geiszler PC, Mesquita-Ribeiro R, Scott D, Layfield R, Serres S. A convenient model of serum-induced reactivity of human astrocytes to investigate astrocyte-derived extracellular vesicles. Front Cell Neurosci 2024; 18:1414142. [PMID: 38915876 PMCID: PMC11195030 DOI: 10.3389/fncel.2024.1414142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 05/17/2024] [Indexed: 06/26/2024] Open
Abstract
Extracellular vesicles (EVs) are secreted by all cells in the CNS, including neurons and astrocytes. EVs are lipid membrane enclosed particles loaded with various bioactive cargoes reflecting the dynamic activities of cells of origin. In contrast to neurons, the specific role of EVs released by astrocytes is less well understood, partly due to the difficulty in maintaining primary astrocyte cultures in a quiescent state. The aim of this study was to establish a human serum-free astrocyte culture system that maintains primary astrocytes in a quiescent state to study the morphology, function, and protein cargoes of astrocyte-derived EVs. Serum-free medium with G5 supplement and serum-supplemented medium with 2% FBS were compared for the culture of commercially available human primary fetal astrocytes. Serum-free astrocytes displayed morphologies similar to in vivo astrocytes, and surprisingly, higher levels of astrocyte markers compared to astrocytes chronically cultured in FBS. In contrast, astrocyte and inflammatory markers in serum-free astrocytes were upregulated 24 h after either acute 2% FBS or cytokine exposure, confirming their capacity to become reactive. Importantly, this suggests that distinct signaling pathways are involved in acute and chronic astrocyte reactivity. Despite having a similar morphology, chronically serum-cultured astrocyte-derived EVs (ADEVs) were smaller in size compared to serum-free ADEVs and could reactivate serum-free astrocytes. Proteomic analysis identified distinct protein datasets for both types of ADEVs with enrichment of complement and coagulation cascades for chronically serum-cultured astrocyte-derived EVs, offering insights into their roles in the CNS. Collectively, these results suggest that human primary astrocytes cultured in serum-free medium bear similarities with in vivo quiescent astrocytes and the addition of serum induces multiple morphological and transcriptional changes that are specific to human reactive astrocytes and their ADEVs. Thus, more emphasis should be made on using multiple structural, molecular, and functional parameters when evaluating ADEVs as biomarkers of astrocyte health.
Collapse
Affiliation(s)
- Katherine E. White
- School of Life Sciences, University of Nottingham, Nottingham, United Kingdom
| | - Hannah L. Bailey
- School of Life Sciences, University of Nottingham, Nottingham, United Kingdom
| | - Barry S. Shaw
- School of Life Sciences, University of Nottingham, Nottingham, United Kingdom
| | | | | | - Daniel Scott
- School of Life Sciences, University of Nottingham, Nottingham, United Kingdom
| | - Robert Layfield
- School of Life Sciences, University of Nottingham, Nottingham, United Kingdom
| | - Sébastien Serres
- School of Life Sciences, University of Nottingham, Nottingham, United Kingdom
- The David Greenfield Human Physiology Unit, University of Nottingham, Nottingham, United Kingdom
| |
Collapse
|
1055
|
Wang L, Zhang G, Gao Y, Dai T, Yu J, Liu Y, Bao H, She J, Hou Y, Kong L, Cai B. Extracellular Vesicles Derived from Neutrophils Accelerate Bone Regeneration by Promoting Osteogenic Differentiation of BMSCs. ACS Biomater Sci Eng 2024; 10:3868-3882. [PMID: 38703236 PMCID: PMC11167592 DOI: 10.1021/acsbiomaterials.4c00106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 04/22/2024] [Accepted: 04/30/2024] [Indexed: 05/06/2024]
Abstract
The reconstruction of bone defects has been associated with severe challenges worldwide. Nowadays, bone marrow mesenchymal stem cell (BMSC)-based cell sheets have rendered this approach a promising way to facilitate osteogenic regeneration in vivo. Extracellular vesicles (EVs) play an essential role in intercellular communication and execution of various biological functions and are often employed as an ideal natural endogenous nanomedicine for restoring the structure and functions of damaged tissues. The perception of polymorphonuclear leukocytes (neutrophils, PMNs) as indiscriminate killer cells is gradually changing, with new evidence suggesting a role for these cells in tissue repair and regeneration, particularly in the context of bone healing. However, the role of EVs derived from PMNs (PMN-EVs) in bone regeneration remains largely unknown, with limited research being conducted on this aspect. In the current study, we investigated the effects of PMN-EVs on BMSCs and the underlying molecular mechanisms as well as the potential application of PMN-EVs in bone regeneration. Toward this end, BMSC-based cell sheets with integrated PMN-EVs (BS@PMN-EVs) were developed for bone defect regeneration. PMN-EVs were found to significantly enhance the proliferation and osteogenic differentiation of BMSCs in vitro. Furthermore, BS@PMN-EVs were found to significantly accelerate bone regeneration in vivo by enhancing the maturation of the newly formed bone in rat calvarial defects; this is likely attributable to the effect of PMN-EVs in promoting the expression of key osteogenic proteins such as SOD2 and GJA1 in BMSCs. In conclusion, our findings demonstrate the crucial role of PMN-EVs in promoting the osteogenic differentiation of BMSCs during bone regeneration. Furthermore, this study proposes a novel strategy for enhancing bone repair and regeneration via the integration of PMN-EVs with BMSC-based cell sheets.
Collapse
Affiliation(s)
- Le Wang
- State
Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration,
National Clinical Research Center for Oral Diseases, Shaanxi Clinical
Research Center for Oral Diseases, Department of Oral and Maxillofacial
Surgery, School of Stomatology, The Fourth
Military Medical University, Xi’an 710032, China
| | - Guanhua Zhang
- State
Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration,
National Clinical Research Center for Oral Diseases, Shaanxi Clinical
Research Center for Oral Diseases, Department of Oral Implants, School
of Stomatology, The Fourth Military Medical
University, Xi’an 710032, China
| | - Ye Gao
- State
Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration,
National Clinical Research Center for Oral Diseases, Shaanxi Clinical
Research Center for Oral Diseases, Department of Oral and Maxillofacial
Surgery, School of Stomatology, The Fourth
Military Medical University, Xi’an 710032, China
| | - Taiqiang Dai
- State
Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration,
National Clinical Research Center for Oral Diseases, Shaanxi Clinical
Research Center for Oral Diseases, Department of Oral and Maxillofacial
Surgery, School of Stomatology, The Fourth
Military Medical University, Xi’an 710032, China
| | - Jie Yu
- State
Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration,
National Clinical Research Center for Oral Diseases, Shaanxi Clinical
Research Center for Oral Diseases, Department of Oral and Maxillofacial
Surgery, School of Stomatology, The Fourth
Military Medical University, Xi’an 710032, China
| | - Ya Liu
- State
Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration,
National Clinical Research Center for Oral Diseases, Shaanxi Clinical
Research Center for Oral Diseases, Department of Oral and Maxillofacial
Surgery, School of Stomatology, The Fourth
Military Medical University, Xi’an 710032, China
- College
of Life Sciences, Northwest University, Xi’an 710069, China
| | - Han Bao
- State
Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration,
National Clinical Research Center for Oral Diseases, Shaanxi Clinical
Research Center for Oral Diseases, Department of Oral and Maxillofacial
Surgery, School of Stomatology, The Fourth
Military Medical University, Xi’an 710032, China
| | - Jianzhen She
- State
Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration,
National Clinical Research Center for Oral Diseases, Shaanxi Clinical
Research Center for Oral Diseases, Department of Oral and Maxillofacial
Surgery, School of Stomatology, The Fourth
Military Medical University, Xi’an 710032, China
| | - Yan Hou
- State
Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration,
National Clinical Research Center for Oral Diseases, Shaanxi Clinical
Research Center for Oral Diseases, Department of Oral and Maxillofacial
Surgery, School of Stomatology, The Fourth
Military Medical University, Xi’an 710032, China
| | - Liang Kong
- State
Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration,
National Clinical Research Center for Oral Diseases, Shaanxi Clinical
Research Center for Oral Diseases, Department of Oral and Maxillofacial
Surgery, School of Stomatology, The Fourth
Military Medical University, Xi’an 710032, China
| | - Bolei Cai
- State
Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration,
National Clinical Research Center for Oral Diseases, Shaanxi Clinical
Research Center for Oral Diseases, Department of Oral and Maxillofacial
Surgery, School of Stomatology, The Fourth
Military Medical University, Xi’an 710032, China
| |
Collapse
|
1056
|
Jank L, Kesharwani A, Ryu T, Joshi D, Ladakis DC, Smith MD, Singh S, Arab T, Witwer KW, Calabresi PA, Na CH, Bhargava P. Characterization of spinal cord tissue-derived extracellular vesicles in neuroinflammation. J Neuroinflammation 2024; 21:154. [PMID: 38851724 PMCID: PMC11162576 DOI: 10.1186/s12974-024-03147-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 05/30/2024] [Indexed: 06/10/2024] Open
Abstract
Extracellular vesicles (EVs) are released by all cells, can cross the blood-brain barrier, and have been shown to play an important role in cellular communication, substance shuttling, and immune modulation. In recent years EVs have shifted into focus in multiple sclerosis (MS) research as potential plasma biomarkers and therapeutic vehicles. Yet little is known about the disease-associated changes in EVs in the central nervous system (CNS). To address this gap, we characterized the physical and proteomic changes of mouse spinal cord-derived EVs before and at 16 and 25 days after the induction of experimental autoimmune encephalomyelitis (EAE), a neuroinflammatory model of MS. Using various bioinformatic tools, we found changes in inflammatory, glial, and synaptic proteins and pathways, as well as a shift in the predicted contribution of immune and glial cell types over time. These results show that EVs provide snapshots of crucial disease processes such as CNS-compartmentalized inflammation, re/de-myelination, and synaptic pathology, and might also mediate these processes. Additionally, inflammatory plasma EV biomarkers previously identified in people with MS were also altered in EAE spinal cord EVs, suggesting commonalities of EV-related pathological processes during EAE and MS and overlap of EV proteomic changes between CNS and circulating EVs.
Collapse
Affiliation(s)
- Larissa Jank
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ajay Kesharwani
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Taekyung Ryu
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Deepika Joshi
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Dimitrios C Ladakis
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Matthew D Smith
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Saumitra Singh
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Tanina Arab
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Kenneth W Witwer
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Peter A Calabresi
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Chan-Hyun Na
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Pavan Bhargava
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
1057
|
Couse AD, Cox-Vazquez SJ, Ghatak S, Trinidad JC, Clemmer DE. Delineating Bovine Milk Derived Microvesicles from Exosomes Using Proteomics. J Proteome Res 2024; 23:2288-2297. [PMID: 38805445 PMCID: PMC12161284 DOI: 10.1021/acs.jproteome.4c00352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/30/2024]
Abstract
In the work presented herein, a simple serial-pelleting purification strategy combined with a mass spectrometry-based proteomics analysis was developed as a means of discerning differences in extracellular vesicle (EV) populations found in bovine milk samples. A sequence of ultracentrifugation speeds was used to generate changes in the abundances of EV populations, allowing for the identification of associated proteins. A metric was developed to determine the relative abundances of proteins in large EVs (>200 nm) and small EVs (<200 nm). Of the 476 proteins consistently found in this study, 340 are associated with vesicular components. Of these, 156 were heavily enriched in large EVs, 155 shared between large and small EVs, and 29 heavily enriched in small EVs. Additionally, out of 68 proteins annotated as exosome proteins, 32 were enriched in large EVs, 27 shared between large and small EVs, 5 enriched in small EVs, and 7 were found to be nonvesicular contaminant proteins. The top correlated proteins in the small EV group were predominantly membrane-bound proteins, whereas the top correlated proteins in the large EV group were mostly cytosolic enzymes for molecular processing. This method provides a means of assessing the origins of vesicle components and provides new potential marker proteins within discrete vesicle populations.
Collapse
Affiliation(s)
- Andrew D. Couse
- Department of Chemistry, Indiana University Bloomington, Bloomington, Indiana 47405, United States
| | - Sarah J. Cox-Vazquez
- Department of Chemistry, Indiana University Bloomington, Bloomington, Indiana 47405, United States
| | - Subhadip Ghatak
- McGowan Institute of Regenerative Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania 15219, United States
| | - Jonathan C. Trinidad
- Department of Chemistry, Indiana University Bloomington, Bloomington, Indiana 47405, United States
| | - David E. Clemmer
- Department of Chemistry, Indiana University Bloomington, Bloomington, Indiana 47405, United States
| |
Collapse
|
1058
|
Yang S, Sun Y, Yan C. Recent advances in the use of extracellular vesicles from adipose-derived stem cells for regenerative medical therapeutics. J Nanobiotechnology 2024; 22:316. [PMID: 38844939 PMCID: PMC11157933 DOI: 10.1186/s12951-024-02603-4] [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: 03/21/2024] [Accepted: 05/28/2024] [Indexed: 06/09/2024] Open
Abstract
Adipose-derived stem cells (ADSCs) are a subset of mesenchymal stem cells (MSCs) isolated from adipose tissue. They possess remarkable properties, including multipotency, self-renewal, and easy clinical availability. ADSCs are also capable of promoting tissue regeneration through the secretion of various cytokines, factors, and extracellular vesicles (EVs). ADSC-derived EVs (ADSC-EVs) act as intercellular signaling mediators that encapsulate a range of biomolecules. These EVs have been found to mediate the therapeutic activities of donor cells by promoting the proliferation and migration of effector cells, facilitating angiogenesis, modulating immunity, and performing other specific functions in different tissues. Compared to the donor cells themselves, ADSC-EVs offer advantages such as fewer safety concerns and more convenient transportation and storage for clinical application. As a result, these EVs have received significant attention as cell-free therapeutic agents with potential future application in regenerative medicine. In this review, we focus on recent research progress regarding regenerative medical use of ADSC-EVs across various medical conditions, including wound healing, chronic limb ischemia, angiogenesis, myocardial infarction, diabetic nephropathy, fat graft survival, bone regeneration, cartilage regeneration, tendinopathy and tendon healing, peripheral nerve regeneration, and acute lung injury, among others. We also discuss the underlying mechanisms responsible for inducing these therapeutic effects. We believe that deciphering the biological properties, therapeutic effects, and underlying mechanisms associated with ADSC-EVs will provide a foundation for developing a novel therapeutic approach in regenerative medicine.
Collapse
Affiliation(s)
- Song Yang
- Institute of Life Sciences, Chongqing Medical University, Chongqing, 400016, People's Republic of China.
| | - Yiran Sun
- School of Pharmacy, Chengdu Medical College, Chengdu, 610500, People's Republic of China.
| | - Chenchen Yan
- School of Pharmacy, Chengdu Medical College, Chengdu, 610500, People's Republic of China
| |
Collapse
|
1059
|
Guerricchio L, Barile L, Bollini S. Evolving Strategies for Extracellular Vesicles as Future Cardiac Therapeutics: From Macro- to Nano-Applications. Int J Mol Sci 2024; 25:6187. [PMID: 38892376 PMCID: PMC11173118 DOI: 10.3390/ijms25116187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 05/28/2024] [Accepted: 05/31/2024] [Indexed: 06/21/2024] Open
Abstract
Cardiovascular disease represents the foremost cause of mortality and morbidity worldwide, with a steadily increasing incidence due to the growth of the ageing population. Cardiac dysfunction leading to heart failure may arise from acute myocardial infarction (MI) as well as inflammatory- and cancer-related chronic cardiomyopathy. Despite pharmacological progress, effective cardiac repair represents an unmet clinical need, with heart transplantation being the only option for end-stage heart failure. The functional profiling of the biological activity of extracellular vesicles (EVs) has recently attracted increasing interest in the field of translational research for cardiac regenerative medicine. The cardioprotective and cardioactive potential of human progenitor stem/cell-derived EVs has been reported in several preclinical studies, and EVs have been suggested as promising paracrine therapy candidates for future clinical translation. Nevertheless, some compelling aspects must be properly addressed, including optimizing delivery strategies to meet patient needs and enhancing targeting specificity to the cardiac tissue. Therefore, in this review, we will discuss the most relevant aspects of the therapeutic potential of EVs released by human progenitors for cardiovascular disease, with a specific focus on the strategies that have been recently implemented to improve myocardial targeting and administration routes.
Collapse
Affiliation(s)
- Laura Guerricchio
- Biology Unit, Department of Experimental Medicine (DIMES), University of Genova, 16132 Genova, Italy;
| | - Lucio Barile
- Cardiovascular Theranostics, Istituto Cardiocentro Ticino, Laboratories for Translational Research, Ente Ospedaliero Cantonale, CH-6500 Bellinzona, Switzerland;
- Euler Institute, Faculty of Biomedical Sciences, Università della Svizzera Italiana, CH-6900 Lugano, Switzerland
| | - Sveva Bollini
- Biology Unit, Department of Experimental Medicine (DIMES), University of Genova, 16132 Genova, Italy;
- Cellular Oncology Unit, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy
| |
Collapse
|
1060
|
Calzoni E, Cerrotti G, Sagini K, Delo F, Buratta S, Pellegrino RM, Alabed HBR, Fratini F, Emiliani C, Urbanelli L. Evidence of Lysosomal β-Hexosaminidase Enzymatic Activity Associated with Extracellular Vesicles: Potential Applications for the Correction of Sandhoff Disease. J Funct Biomater 2024; 15:153. [PMID: 38921527 PMCID: PMC11204914 DOI: 10.3390/jfb15060153] [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: 04/28/2024] [Revised: 05/21/2024] [Accepted: 05/30/2024] [Indexed: 06/27/2024] Open
Abstract
Extracellular vesicles (EVs) can be isolated from biological fluids and cell culture medium. Their nanometric dimension, relative stability, and biocompatibility have raised considerable interest for their therapeutic use as delivery vehicles of macromolecules, namely nucleic acids and proteins. Deficiency in lysosomal enzymes and associated proteins is at the basis of a group of genetic diseases known as lysosomal storage disorders (LSDs), characterized by the accumulation of undigested substrates into lysosomes. Among them, GM2 gangliosidoses are due to a deficiency in the activity of lysosomal enzyme β-hexosaminidase, leading to the accumulation of the GM2 ganglioside and severe neurological symptoms. Current therapeutic approaches, including enzyme replacement therapy (ERT), have proven unable to significantly treat these conditions. Here, we provide evidence that the lysosomal β-hexosaminidase enzyme is associated with EVs released by HEK cells and that the EV-associated activity can be increased by overexpressing the α-subunit of β-hexosaminidase. The delivery of EVs to β-hexosaminidase-deficient fibroblasts results in a partial cross-correction of the enzymatic defect. Overall findings indicate that EVs could be a source of β-hexosaminidase that is potentially exploitable for developing therapeutic approaches for currently untreatable LSDs.
Collapse
Affiliation(s)
- Eleonora Calzoni
- Department of Chemistry, Biology and Biotechnology, University of Perugia, 06100 Perugia, Italy; (E.C.); (G.C.); (K.S.); (F.D.); (S.B.); (R.M.P.); (H.B.R.A.); (C.E.)
| | - Giada Cerrotti
- Department of Chemistry, Biology and Biotechnology, University of Perugia, 06100 Perugia, Italy; (E.C.); (G.C.); (K.S.); (F.D.); (S.B.); (R.M.P.); (H.B.R.A.); (C.E.)
| | - Krizia Sagini
- Department of Chemistry, Biology and Biotechnology, University of Perugia, 06100 Perugia, Italy; (E.C.); (G.C.); (K.S.); (F.D.); (S.B.); (R.M.P.); (H.B.R.A.); (C.E.)
| | - Federica Delo
- Department of Chemistry, Biology and Biotechnology, University of Perugia, 06100 Perugia, Italy; (E.C.); (G.C.); (K.S.); (F.D.); (S.B.); (R.M.P.); (H.B.R.A.); (C.E.)
| | - Sandra Buratta
- Department of Chemistry, Biology and Biotechnology, University of Perugia, 06100 Perugia, Italy; (E.C.); (G.C.); (K.S.); (F.D.); (S.B.); (R.M.P.); (H.B.R.A.); (C.E.)
- Centro di Eccellenza sui Materiali Innovativi Nanostrutturati (CEMIN), University of Perugia, 06123 Perugia, Italy
| | - Roberto Maria Pellegrino
- Department of Chemistry, Biology and Biotechnology, University of Perugia, 06100 Perugia, Italy; (E.C.); (G.C.); (K.S.); (F.D.); (S.B.); (R.M.P.); (H.B.R.A.); (C.E.)
| | - Husam B. R. Alabed
- Department of Chemistry, Biology and Biotechnology, University of Perugia, 06100 Perugia, Italy; (E.C.); (G.C.); (K.S.); (F.D.); (S.B.); (R.M.P.); (H.B.R.A.); (C.E.)
| | | | - Carla Emiliani
- Department of Chemistry, Biology and Biotechnology, University of Perugia, 06100 Perugia, Italy; (E.C.); (G.C.); (K.S.); (F.D.); (S.B.); (R.M.P.); (H.B.R.A.); (C.E.)
- Centro di Eccellenza sui Materiali Innovativi Nanostrutturati (CEMIN), University of Perugia, 06123 Perugia, Italy
| | - Lorena Urbanelli
- Department of Chemistry, Biology and Biotechnology, University of Perugia, 06100 Perugia, Italy; (E.C.); (G.C.); (K.S.); (F.D.); (S.B.); (R.M.P.); (H.B.R.A.); (C.E.)
- Centro di Eccellenza sui Materiali Innovativi Nanostrutturati (CEMIN), University of Perugia, 06123 Perugia, Italy
| |
Collapse
|
1061
|
Garrett NR, Pink RC, Lawson C. Contribution of Extracellular Particles Isolated from Morus sp. (Mulberry) Fruit to Their Reported Protective Health Benefits: An In Vitro Study. Int J Mol Sci 2024; 25:6177. [PMID: 38892365 PMCID: PMC11173249 DOI: 10.3390/ijms25116177] [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: 05/01/2024] [Revised: 05/29/2024] [Accepted: 05/31/2024] [Indexed: 06/21/2024] Open
Abstract
Morus sp. (mulberry) has a long tradition of use as a medicinal treatment, including for cardiovascular disease and type 2 diabetes, being shown to have antioxidant properties and to promote wound healing. Extracellular vesicles (EVs) are sub-micron, membrane-enclosed particles that were first identified in mammalian bodily fluids. EV-like particles have been described in plants (PDVs) and shown to have similar characteristics to mammalian EVs. We hypothesised that some of the health benefits previously attributed to the fruit of Morus sp. could be due to the release of PDVs. We isolated PDVs from Morus nigra and Morus alba via ultracentrifugation and incubated THP-1 monocytes, differentiated THP-1 macrophages, or HMEC-1 endothelial cells with pro-oxidant compounds DMNQ (THP-1) and glucose oxidase (HMEC-1) or lipopolysaccharide (LPS) in the presence of different fractions of mulberry EVs. Mulberry EVs augmented ROS production with DMNQ in THP-1 and caused the downregulation of ROS in HMEC-1. Mulberry EVs increased LPS-induced IL-1β secretion but reduced CCL2 and TGF-β secretion in THP-1 macrophages. In scratch wound assays, mulberry EVs inhibited HMEC-1 migration but increased proliferation in both low and high serum conditions, suggesting that they have opposing effects in these two important aspects of wound healing. One of the limitations of plant-derived therapeutics has been overcoming the low bioavailability of isolated compounds. We propose that PDVs could provide the link between physiological dose and therapeutic benefit by protecting plant active compounds in the GIT as well as potentially delivering genetic material or proteins that contribute to previously observed health benefits.
Collapse
Affiliation(s)
- Neve R. Garrett
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London NW1 0TU, UK;
| | - Ryan C. Pink
- Department of Biological and Medical Sciences, Oxford Brookes University, Oxford OX3 0BP, UK;
| | - Charlotte Lawson
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London NW1 0TU, UK;
- School of Pharmacy and Biomedical Sciences, University of Central Lancashire, Preston PR1 2HE, UK
| |
Collapse
|
1062
|
Oberholster L, Du Pasquier R, Mathias A. Exploring the role of brain-derived extracellular vesicles in viral infections: from pathological insights to biomarker potential. Front Cell Infect Microbiol 2024; 14:1423394. [PMID: 38887492 PMCID: PMC11181307 DOI: 10.3389/fcimb.2024.1423394] [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: 04/25/2024] [Accepted: 05/21/2024] [Indexed: 06/20/2024] Open
Abstract
Extracellular vesicles (EVs) are membrane-bound vesicles secreted by all cell types that play a central role in cell-to-cell communication. Since these vesicles serve as vehicles of cellular content (nucleic acids, proteins and lipids) with the potential to cross biological barriers, they represent a novel attractive window into an otherwise inaccessible organ, such as the brain. The composition of EVs is cell-type specific and mirrors the physiological condition of the cell-of-origin. Consequently, during viral infection, EVs undergo significant changes in their content and morphology, thereby reflecting alterations in the cellular state. Here, we briefly summarize the potential of brain-derived EVs as a lens into viral infection in the central nervous system, thereby: 1) uncovering underlying pathophysiological processes at play and 2) serving as liquid biopsies of the brain, representing a non-invasive source of biomarkers for monitoring disease activity. Although translating the potential of EVs from research to diagnosis poses complexities, characterizing brain-derived EVs in the context of viral infections holds promise to enhance diagnostic and therapeutic strategies, offering new avenues for managing infectious neurological diseases.
Collapse
Affiliation(s)
- Larise Oberholster
- Laboratory of Neuroimmunology, Service of Neurology, Department of Clinical Neurosciences, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland
| | - Renaud Du Pasquier
- Laboratory of Neuroimmunology, Service of Neurology, Department of Clinical Neurosciences, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland
- Service of Neurology, Department of Clinical Neurosciences, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland
| | - Amandine Mathias
- Laboratory of Neuroimmunology, Service of Neurology, Department of Clinical Neurosciences, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
1063
|
Chen X, Hu X, Liu T. Development of liquid biopsy in detection and screening of pancreatic cancer. Front Oncol 2024; 14:1415260. [PMID: 38887233 PMCID: PMC11180763 DOI: 10.3389/fonc.2024.1415260] [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: 04/12/2024] [Accepted: 05/14/2024] [Indexed: 06/20/2024] Open
Abstract
Pancreatic cancer is a highly lethal malignant tumor, which has the characteristics of occult onset, low early diagnosis rate, rapid development and poor prognosis. The reason for the high mortality is partly that pancreatic cancer is usually found in the late stage and missed the best opportunity for surgical resection. As a promising detection technology, liquid biopsy has the advantages of non-invasive, real-time and repeatable. In recent years, the continuous development of liquid biopsy has provided a new way for the detection and screening of pancreatic cancer. The update of biomarkers and detection tools has promoted the development of liquid biopsy. Circulating tumor cells (CTCs), circulating tumor DNA (ctDNA), circulating tumor RNA (ctRNA) and extracellular vesicles (EVs) provide many biomarkers for liquid biopsy of pancreatic cancer, and screening tools around them have also been developed. This review aims to report the application of liquid biopsy technology in the detection of pancreatic cancer patients, mainly introduces the biomarkers and some newly developed tools and platforms. We have also considered whether liquid biopsy technology can replace traditional tissue biopsy and the challenges it faces.
Collapse
Affiliation(s)
- Xiangcheng Chen
- Key Laboratory of Antibody Engineering of Guangdong Higher Education Institutes, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, Guangdong, China
| | - Xinyi Hu
- School of The First Clinical Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Tiancai Liu
- Key Laboratory of Antibody Engineering of Guangdong Higher Education Institutes, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
1064
|
Cheng CA. Before Translating Extracellular Vesicles into Personalized Diagnostics and Therapeutics: What We Could Do. Mol Pharm 2024; 21:2625-2636. [PMID: 38771015 DOI: 10.1021/acs.molpharmaceut.4c00185] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Extracellular vesicle (EV) research is rapidly advancing from fundamental science to translational applications in EV-based personalized therapeutics and diagnostics. Yet, fundamental questions persist regarding EV biology and mechanisms, particularly concerning the heterogeneous interactions between EVs and cells. While we have made strides in understanding virus delivery and intracellular vesicle transport, our comprehension of EV trafficking remains limited. EVs are believed to mediate intercellular communication through cargo transfer, but uncertainties persist regarding the occurrence and quantification of EV-cargo delivery within acceptor cells. This ambiguity is crucial to address, given the significant translational impact of EVs on therapeutics and diagnostics. This perspective article does not seek to provide exhaustive recommendations and guidance on EV-related studies, as these are well-articulated in position papers and statements by the International Society for Extracellular Vesicles (ISEV), including the 'Minimum Information for Studies of Extracellular Vesicles' (MISEV) 2014, MISEV2018, and the recent MISEV2023. Instead, recognizing the multilayered heterogeneity of EVs as both a challenge and an opportunity, this perspective emphasizes novel approaches to facilitate our understanding of diverse EV biology, address uncertainties, and leverage this knowledge to advance EV-based personalized diagnostics and therapeutics. Specifically, this perspective synthesizes current insights, identifies opportunities, and highlights exciting technological advancements in ultrasensitive single EV or "digital" profiling developed within the author's multidisciplinary group. These newly developed technologies address technical gaps in dissecting the molecular contents of EV subsets, contributing to the evolution of EVs as next-generation liquid biopsies for diagnostics and providing better quality control for EV-based therapeutics.
Collapse
Affiliation(s)
- Chi-An Cheng
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei 10050, Taiwan
| |
Collapse
|
1065
|
Huang S, Zeng Y, Guo Q, Zou T, Yin ZQ. Small extracellular vesicles of organoid-derived human retinal stem cells remodel Müller cell fate via miRNA: A novel remedy for retinal degeneration. J Control Release 2024; 370:405-420. [PMID: 38663753 DOI: 10.1016/j.jconrel.2024.04.036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Revised: 04/19/2024] [Accepted: 04/22/2024] [Indexed: 05/08/2024]
Abstract
Remodeling retinal Müller glial fate, including gliosis inhibition and pro-reprogramming, represents a crucial avenue for treating degenerative retinal diseases. Stem cell transplantation exerts effects on modulating retinal Müller glial fate. However, the optimized stem cell products and the underlying therapeutic mechanisms need to be investigated. In the present study, we found that retinal progenitor cells from human embryonic stem cell-derived retinal organoids (hERO-RPCs) transferred extracellular vesicles (EVs) into Müller cells following subretinal transplantation into RCS rats. Small EVs from hERO-RPCs (hERO-RPC-sEVs) were collected and were found to delay photoreceptor degeneration and protect retinal function in RCS rats. hERO-RPC-sEVs were taken up by Müller cells both in vivo and in vitro, and inhibited gliosis while promoting early dedifferentiation of Müller cells. We further explored the miRNA profiles of hERO-RPC-sEVs, which suggested a functional signature associated with neuroprotection and development, as well as the regulation of stem cell and glial fate. Mechanistically, hERO-RPC-sEVs might regulate the fate of Müller cells by miRNA-mediated nuclear factor I transcription factors B (NFIB) downregulation. Collectively, our findings offer novel mechanistic insights into stem cell therapy and promote the development of EV-centered therapeutic strategies.
Collapse
Affiliation(s)
- Shudong Huang
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China; Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing 400038, China
| | - Yuxiao Zeng
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China; Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing 400038, China
| | - Qiang Guo
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China; Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing 400038, China
| | - Ting Zou
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China; Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing 400038, China; Department of Ophthalmology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China.
| | - Zheng Qin Yin
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China; Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing 400038, China.
| |
Collapse
|
1066
|
Artuyants A, Guo G, Flinterman M, Middleditch M, Jacob B, Lee K, Vella L, Su H, Wilson M, Eva L, Shelling AN, Blenkiron C. The tumour-derived extracellular vesicle proteome varies by endometrial cancer histology and is confounded by an obesogenic environment. Proteomics 2024; 24:e2300055. [PMID: 38644352 DOI: 10.1002/pmic.202300055] [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: 09/14/2023] [Revised: 04/03/2024] [Accepted: 04/05/2024] [Indexed: 04/23/2024]
Abstract
Endometrial cancer, the most common gynaecological cancer worldwide, is closely linked to obesity and metabolic diseases, particularly in younger women. New circulating biomarkers have the potential to improve diagnosis and treatment selections, which could significantly improve outcomes. Our approach focuses on extracellular vesicle (EV) biomarker discovery by directly profiling the proteome of EVs enriched from frozen biobanked endometrial tumours. We analysed nine tissue samples to compare three clinical subgroups-low BMI (Body Mass Index) Endometrioid, high BMI Endometrioid, and Serous (any BMI)-identifying proteins related to histological subtype, BMI, and shared secreted proteins. Using collagenase digestion and size exclusion chromatography, we successfully enriched generous quantities of EVs (range 204.8-1291.0 µg protein: 1.38 × 1011-1.10 × 1012 particles), characterised by their size (∼150 nm), expression of EV markers (CD63/81), and proposed endometrial cancer markers (L1CAM, ANXA2). Mass spectrometry-based proteomic profiling identified 2075 proteins present in at least one of the 18 samples. Compared to cell lysates, EVs were successfully depleted for mitochondrial and blood proteins and enriched for common EV markers and large secreted proteins. Further analysis highlighted significant differences in EV protein profiles between the high BMI subgroup and others, underlining the impact of comorbidities on the EV secretome. Interestingly, proteins differentially abundant in tissue subgroups were largely not also differential in matched EVs. This research identified secreted proteins known to be involved in endometrial cancer pathophysiology and proposed novel diagnostic biomarkers (EIF6, MUC16, PROM1, SLC26A2).
Collapse
Affiliation(s)
- Anastasiia Artuyants
- Department of Molecular Medicine and Pathology, The University of Auckland, Auckland, New Zealand
- Auckland Cancer Society Research Centre, The University of Auckland, Auckland, New Zealand
| | - George Guo
- Department of Physiology in the School of Medical Sciences, The University of Auckland, Auckland, New Zealand
- Mass Spectrometry Hub, The University of Auckland, Auckland, New Zealand
| | - Marcella Flinterman
- Auckland Cancer Society Research Centre, The University of Auckland, Auckland, New Zealand
| | - Martin Middleditch
- Technical Services, Faculty of Science, The University of Auckland, Auckland, New Zealand
| | - Bincy Jacob
- Centre of eResearch, Faculty of Science, The University of Auckland, Auckland, New Zealand
| | - Kate Lee
- Department of Molecular Medicine and Pathology, The University of Auckland, Auckland, New Zealand
| | - Laura Vella
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia
- Department of Surgery, The Royal Melbourne Hospital, The University of Melbourne, Parkville, Victoria, Australia
| | - Huaqi Su
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia
| | - Michelle Wilson
- Cancer and Blood, Auckland City Hospital, Auckland, New Zealand
- Department of Oncology, The University of Auckland, Auckland, New Zealand
| | - Lois Eva
- Department of Gynaecological Oncology, Auckland City Hospital, Auckland, New Zealand
- Department of Obstetrics and Gynaecology, The University of Auckland, Auckland, New Zealand
| | - Andrew N Shelling
- Department of Obstetrics and Gynaecology, The University of Auckland, Auckland, New Zealand
- Centre for Cancer Research, The University of Auckland, Auckland, New Zealand
| | - Cherie Blenkiron
- Department of Molecular Medicine and Pathology, The University of Auckland, Auckland, New Zealand
- Auckland Cancer Society Research Centre, The University of Auckland, Auckland, New Zealand
| |
Collapse
|
1067
|
Nguyen VVT, Welsh JA, Tertel T, Choo A, van de Wakker SI, Defourny KAY, Giebel B, Vader P, Padmanabhan J, Lim SK, Nolte‐'t Hoen ENM, Verhaar MC, Bostancioglu RB, Zickler AM, Hong JM, Jones JC, EL Andaloussi S, van Balkom BWM, Görgens A. Inter-laboratory multiplex bead-based surface protein profiling of MSC-derived EV preparations identifies MSC-EV surface marker signatures. J Extracell Vesicles 2024; 13:e12463. [PMID: 38868945 PMCID: PMC11170075 DOI: 10.1002/jev2.12463] [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: 10/20/2023] [Revised: 04/15/2024] [Accepted: 05/21/2024] [Indexed: 06/14/2024] Open
Abstract
Mesenchymal stromal cells (MSCs) are promising regenerative therapeutics that primarily exert their effects through secreted extracellular vesicles (EVs). These EVs - being small and non-living - are easier to handle and possess advantages over cellular products. Consequently, the therapeutic potential of MSC-EVs is increasingly investigated. However, due to variations in MSC-EV manufacturing strategies, MSC-EV products should be considered as highly diverse. Moreover, the diverse array of EV characterisation technologies used for MSC-EV characterisation further complicates reliable interlaboratory comparisons of published data. Consequently, this study aimed to establish a common method that can easily be used by various MSC-EV researchers to characterise MSC-EV preparations to facilitate interlaboratory comparisons. To this end, we conducted a comprehensive inter-laboratory assessment using a novel multiplex bead-based EV flow cytometry assay panel. This assessment involved 11 different MSC-EV products from five laboratories with varying MSC sources, culture conditions, and EV preparation methods. Through this assay panel covering a range of mostly MSC-related markers, we identified a set of cell surface markers consistently positive (CD44, CD73 and CD105) or negative (CD11b, CD45 and CD197) on EVs of all explored MSC-EV preparations. Hierarchical clustering analysis revealed distinct surface marker profiles associated with specific preparation processes and laboratory conditions. We propose CD73, CD105 and CD44 as robust positive markers for minimally identifying MSC-derived EVs and CD11b, CD14, CD19, CD45 and CD79 as reliable negative markers. Additionally, we highlight the influence of culture medium components, particularly human platelet lysate, on EV surface marker profiles, underscoring the influence of culture conditions on resulting EV products. This standardisable approach for MSC-EV surface marker profiling offers a tool for routine characterisation of manufactured EV products in pre-clinical and clinical research, enhances the quality control of MSC-EV preparations, and hopefully paves the way for higher consistency and reproducibility in the emerging therapeutic MSC-EV field.
Collapse
Affiliation(s)
| | - Joshua A. Welsh
- Translational Nanobiology Section, Laboratory of Pathology, National Cancer InstituteNational Institutes of HealthBethesdaMarylandUSA
- The Measuring Stick, LtdPeterboroughUK
- Advanced Technology GroupBecton DickinsonSan JoseCaliforniaUSA
| | - Tobias Tertel
- Institute for Transfusion MedicineUniversity Hospital EssenUniversity of Duisburg‐EssenEssenGermany
| | - Andre Choo
- Bioprocessing Technology Institute (BTI)Agency for Science, Technology and Research (A*STAR)SingaporeSingapore
| | - Simonides I. van de Wakker
- Department of Cardiology, Experimental Cardiology LaboratoryUniversity Medical Center Utrecht, Utrecht UniversityUtrechtThe Netherlands
| | - Kyra A. Y. Defourny
- Division of Infectious Diseases & Immunology, Department of Biomolecular Health Sciences, Faculty of Veterinary MedicineUtrecht UniversityUtrechtThe Netherlands
| | - Bernd Giebel
- Institute for Transfusion MedicineUniversity Hospital EssenUniversity of Duisburg‐EssenEssenGermany
| | - Pieter Vader
- Department of Cardiology, Experimental Cardiology LaboratoryUniversity Medical Center Utrecht, Utrecht UniversityUtrechtThe Netherlands
- CDL ResearchUniversity Medical Center Utrecht, Utrecht UniversityUtrechtThe Netherlands
| | - Jayanthi Padmanabhan
- Bioprocessing Technology Institute (BTI)Agency for Science, Technology and Research (A*STAR)SingaporeSingapore
| | - Sai Kiang Lim
- Bioprocessing Technology Institute (BTI)Agency for Science, Technology and Research (A*STAR)SingaporeSingapore
| | - Esther N. M. Nolte‐'t Hoen
- Division of Infectious Diseases & Immunology, Department of Biomolecular Health Sciences, Faculty of Veterinary MedicineUtrecht UniversityUtrechtThe Netherlands
| | | | - R. Beklem Bostancioglu
- Division of Biomolecular and Cellular Medicine, Department of Laboratory MedicineKarolinska InstitutetStockholmSweden
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST)Karolinska University Hospital Huddinge and Karolinska Comprehensive Cancer CenterStockholmSweden
| | - Antje M. Zickler
- Division of Biomolecular and Cellular Medicine, Department of Laboratory MedicineKarolinska InstitutetStockholmSweden
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST)Karolinska University Hospital Huddinge and Karolinska Comprehensive Cancer CenterStockholmSweden
- Karolinska ATMP CenterANA FuturaHuddingeSweden
| | - Jia Mei Hong
- Bioprocessing Technology Institute (BTI)Agency for Science, Technology and Research (A*STAR)SingaporeSingapore
| | - Jennifer C. Jones
- Translational Nanobiology Section, Laboratory of Pathology, National Cancer InstituteNational Institutes of HealthBethesdaMarylandUSA
| | - Samir EL Andaloussi
- Division of Biomolecular and Cellular Medicine, Department of Laboratory MedicineKarolinska InstitutetStockholmSweden
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST)Karolinska University Hospital Huddinge and Karolinska Comprehensive Cancer CenterStockholmSweden
- Karolinska ATMP CenterANA FuturaHuddingeSweden
| | | | - André Görgens
- Institute for Transfusion MedicineUniversity Hospital EssenUniversity of Duisburg‐EssenEssenGermany
- Division of Biomolecular and Cellular Medicine, Department of Laboratory MedicineKarolinska InstitutetStockholmSweden
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST)Karolinska University Hospital Huddinge and Karolinska Comprehensive Cancer CenterStockholmSweden
- Karolinska ATMP CenterANA FuturaHuddingeSweden
| |
Collapse
|
1068
|
Greening DW, Rai A, Simpson RJ. Extracellular vesicles-An omics view. Proteomics 2024; 24:e2400128. [PMID: 38676335 DOI: 10.1002/pmic.202400128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 04/15/2024] [Indexed: 04/28/2024]
Affiliation(s)
- David W Greening
- Molecular Proteomics, Baker Heart and Diabetes Institute, Melbourne, Australia
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Australia
- Department of Cardiovascular Research, Translation and Implementation, La Trobe University, Melbourne, VIC, Australia
- Department of Cardiometabolic Health, University of Melbourne, Melbourne, VIC, Australia
| | - Alin Rai
- Molecular Proteomics, Baker Heart and Diabetes Institute, Melbourne, Australia
- Department of Cardiovascular Research, Translation and Implementation, La Trobe University, Melbourne, VIC, Australia
| | - Richard J Simpson
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Australia
| |
Collapse
|
1069
|
Tati V, Mitra S, Basu S, Shukla S. Bone marrow mesenchymal stem cell-derived extracellular vesicles promote corneal epithelial repair and suppress apoptosis via modulation of Caspase-3 in vitro. FEBS Open Bio 2024; 14:968-982. [PMID: 38684330 PMCID: PMC11494918 DOI: 10.1002/2211-5463.13804] [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: 11/03/2023] [Revised: 02/21/2024] [Accepted: 04/10/2024] [Indexed: 05/02/2024] Open
Abstract
Corneal injuries are the major cause of blindness and visual impairment. Available treatments are limited by their efficacy and side effects. Mesenchymal stem cell-derived extracellular vesicles are presumed as functional equivalents and potential candidates for cell-free therapy. This study reports isolation and characterization of extracellular vesicles from human bone marrow mesenchymal stem cells and evaluates their role in mediating epithelial repair and apoptosis in cultured corneal epithelial cells through scratch assay, PCR, immunofluorescence, and flow cytometry in vitro. The isolated extracellular vesicles were spherical, < 150 nm in diameter, and characterized as CD9+, CD63+, CD81+, TSG101+, and Calnexin-. Further, these vesicles promoted corneal epithelial repair by enhancing proliferation and suppressed apoptosis by regulating the expression of BAD, P53, BCL-2, and cleaved CASPASE-3. Thus, our results suggest that BM-MSC-EVs might have the potential to be used for the treatment of injury-induced corneal epithelial defects. Clinical translation of this work would require further investigations.
Collapse
Affiliation(s)
- Vasudeva Tati
- Prof. Brien Holden Eye Research Centre, Hyderabad Eye Research FoundationL V Prasad Eye InstituteHyderabadIndia
- Sudhakar and Sreekanth Ravi Stem Cell Biology Laboratory, Centre for Ocular RegenerationL V Prasad Eye InstituteHyderabadIndia
| | - Sreya Mitra
- Prof. Brien Holden Eye Research Centre, Hyderabad Eye Research FoundationL V Prasad Eye InstituteHyderabadIndia
- Sudhakar and Sreekanth Ravi Stem Cell Biology Laboratory, Centre for Ocular RegenerationL V Prasad Eye InstituteHyderabadIndia
| | - Sayan Basu
- Prof. Brien Holden Eye Research Centre, Hyderabad Eye Research FoundationL V Prasad Eye InstituteHyderabadIndia
- Sudhakar and Sreekanth Ravi Stem Cell Biology Laboratory, Centre for Ocular RegenerationL V Prasad Eye InstituteHyderabadIndia
- Shantilal Shanghvi Cornea Institute, L V Prasad Eye InstituteHyderabadIndia
| | - Sachin Shukla
- Prof. Brien Holden Eye Research Centre, Hyderabad Eye Research FoundationL V Prasad Eye InstituteHyderabadIndia
- Sudhakar and Sreekanth Ravi Stem Cell Biology Laboratory, Centre for Ocular RegenerationL V Prasad Eye InstituteHyderabadIndia
| |
Collapse
|
1070
|
Huang D, Kidd JM, Zou Y, Wu X, Li N, Gehr TWB, Li PL, Li G. Podocyte-specific silencing of acid sphingomyelinase gene to abrogate hyperhomocysteinemia-induced NLRP3 inflammasome activation and glomerular inflammation. Am J Physiol Renal Physiol 2024; 326:F988-F1003. [PMID: 38634138 PMCID: PMC11380990 DOI: 10.1152/ajprenal.00195.2023] [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: 07/13/2023] [Revised: 04/11/2024] [Accepted: 04/16/2024] [Indexed: 04/19/2024] Open
Abstract
Acid sphingomyelinase (ASM) has been reported to increase tissue ceramide and thereby mediate hyperhomocysteinemia (hHcy)-induced glomerular nucleotide-binding oligomerization domain-like receptor containing pyrin domain 3 (NLRP3) inflammasome activation, inflammation, and sclerosis. In the present study, we tested whether somatic podocyte-specific silencing of Smpd1 gene (mouse ASM gene code) attenuates hHcy-induced NLRP3 inflammasome activation and associated extracellular vesicle (EV) release in podocytes and thereby suppresses glomerular inflammatory response and injury. In vivo, somatic podocyte-specific Smpd1 gene silencing almost blocked hHcy-induced glomerular NLRP3 inflammasome activation in Podocre (podocyte-specific expression of cre recombinase) mice compared with control littermates. By nanoparticle tracking analysis (NTA), floxed Smpd1 shRNA transfection was found to abrogate hHcy-induced elevation of urinary EV excretion in Podocre mice. In addition, Smpd1 gene silencing in podocytes prevented hHcy-induced immune cell infiltration into glomeruli, proteinuria, and glomerular sclerosis in Podocre mice. Such protective effects of podocyte-specific Smpd1 gene silencing were mimicked by global knockout of Smpd1 gene in Smpd1-/- mice. On the contrary, podocyte-specific Smpd1 gene overexpression exaggerated hHcy-induced glomerular pathological changes in Smpd1trg/Podocre (podocyte-specific Smpd1 gene overexpression) mice, which were significantly attenuated by transfection of floxed Smpd1 shRNA. In cell studies, we also confirmed that Smpd1 gene knockout or silencing prevented homocysteine (Hcy)-induced elevation of EV release in the primary cultures of podocyte isolated from Smpd1-/- mice or podocytes of Podocre mice transfected with floxed Smpd1 shRNA compared with WT/WT podocytes. Smpd1 gene overexpression amplified Hcy-induced EV secretion from podocytes of Smpd1trg/Podocre mice, which was remarkably attenuated by transfection of floxed Smpd1 shRNA. Mechanistically, Hcy-induced elevation of EV release from podocytes was blocked by ASM inhibitor (amitriptyline, AMI), but not by NLRP3 inflammasome inhibitors (MCC950 and glycyrrhizin, GLY). Super-resolution microscopy also showed that ASM inhibitor, but not NLRP3 inflammasome inhibitors, prevented the inhibition of lysosome-multivesicular body interaction by Hcy in podocytes. Moreover, we found that podocyte-derived inflammatory EVs (released from podocytes treated with Hcy) induced podocyte injury, which was exaggerated by T cell coculture. Interstitial infusion of inflammatory EVs into renal cortex induced glomerular injury and immune cell infiltration. In conclusion, our findings suggest that ASM in podocytes plays a crucial role in the control of NLRP3 inflammasome activation and inflammatory EV release during hHcy and that the development of podocyte-specific ASM inhibition or Smpd1 gene silencing may be a novel therapeutic strategy for treatment of hHcy-induced glomerular disease with minimized side effect.NEW & NOTEWORTHY In the present study, we tested whether podocyte-specific silencing of Smpd1 gene attenuates hyperhomocysteinemia (hHcy)-induced nucleotide-binding oligomerization domain-like receptor containing pyrin domain 3 (NLRP3) inflammasome activation and associated inflammatory extracellular vesicle (EV) release in podocytes and thereby suppresses glomerular inflammatory response and injury. Our findings suggest that acid sphingomyelinase (ASM) in podocytes plays a crucial role in the control of NLRP3 inflammasome activation and inflammatory EV release during hHcy. Based on our findings, it is anticipated that the development of podocyte-specific ASM inhibition or Smpd1 gene silencing may be a novel therapeutic strategy for treatment of hHcy-induced glomerular disease with minimized side effects.
Collapse
Affiliation(s)
- Dandan Huang
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, Virginia, United States
| | - Jason M Kidd
- Division of Nephrology, School of Medicine, Virginia Commonwealth University, Richmond, Virginia, United States
| | - Yao Zou
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, Virginia, United States
| | - Xiaoyuan Wu
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, Virginia, United States
| | - Ningjun Li
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, Virginia, United States
| | - Todd W B Gehr
- Division of Nephrology, School of Medicine, Virginia Commonwealth University, Richmond, Virginia, United States
| | - Pin-Lan Li
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, Virginia, United States
| | - Guangbi Li
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, Virginia, United States
| |
Collapse
|
1071
|
Mason JD, Marks E, Fan S, McCormick K, Wilson C, Harris AL, Hamdy FC, Cunningham C, Goberdhan DCI. Stress-induced Rab11a-exosomes induce amphiregulin-mediated cetuximab resistance in colorectal cancer. J Extracell Vesicles 2024; 13:e12465. [PMID: 38887984 PMCID: PMC11184284 DOI: 10.1002/jev2.12465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 04/28/2024] [Accepted: 05/22/2024] [Indexed: 06/20/2024] Open
Abstract
Exosomes are secreted vesicles made intracellularly in the endosomal system. We have previously shown that exosomes are not only made in late endosomes, but also in recycling endosomes marked by the monomeric G-protein Rab11a. These vesicles, termed Rab11a-exosomes, are preferentially secreted under nutrient stress from several cancer cell types, including HCT116 colorectal cancer (CRC) cells. HCT116 Rab11a-exosomes have particularly potent signalling activities, some mediated by the epidermal growth factor receptor (EGFR) ligand, amphiregulin (AREG). Mutant activating forms of KRAS, a downstream target of EGFR, are often found in advanced CRC. When absent, monoclonal antibodies, such as cetuximab, which target the EGFR and block the effects of EGFR ligands, such as AREG, can be administered. Patients, however, inevitably develop resistance to cetuximab, either by acquiring KRAS mutations or via non-genetic microenvironmental changes. Here we show that nutrient stress in several CRC cell lines causes the release of AREG-carrying Rab11a-exosomes. We demonstrate that while soluble AREG has no effect, much lower levels of AREG bound to Rab11a-exosomes from cetuximab-resistant KRAS-mutant HCT116 cells, can suppress the effects of cetuximab on KRAS-wild type Caco-2 CRC cells. Using neutralising anti-AREG antibodies and an intracellular EGFR kinase inhibitor, we show that this effect is mediated via AREG activation of EGFR, and not transfer of activated KRAS. Therefore, presentation of AREG on Rab11a-exosomes affects its ability to compete with cetuximab. We propose that this Rab11a-exosome-mediated mechanism contributes to the establishment of resistance in cetuximab-sensitive cells and may explain why in cetuximab-resistant tumours only some cells carry mutant KRAS.
Collapse
Affiliation(s)
- John D. Mason
- Department of Physiology, Anatomy and GeneticsUniversity of OxfordOxfordUK
| | - Ewan Marks
- Department of Physiology, Anatomy and GeneticsUniversity of OxfordOxfordUK
| | - Shih‐Jung Fan
- Department of Physiology, Anatomy and GeneticsUniversity of OxfordOxfordUK
- Department of Life SciencesNational Central UniversityTaoyuan CityTaiwan
| | - Kristie McCormick
- Department of Physiology, Anatomy and GeneticsUniversity of OxfordOxfordUK
| | - Clive Wilson
- Department of Physiology, Anatomy and GeneticsUniversity of OxfordOxfordUK
| | - Adrian L. Harris
- Department of Oncology, Weatherall Institute of Molecular MedicineUniversity of OxfordOxfordUK
| | - Freddie C. Hamdy
- Nuffield Department of Surgical SciencesUniversity of Oxford, John Radcliffe HospitalOxfordUK
| | - Chris Cunningham
- Nuffield Department of Surgical SciencesUniversity of Oxford, John Radcliffe HospitalOxfordUK
| | | |
Collapse
|
1072
|
Manni G, Gargaro M, Ricciuti D, Fontana S, Padiglioni E, Cipolloni M, Mazza T, Rosati J, di Veroli A, Mencarelli G, Pieroni B, Silva Barcelos EC, Scalisi G, Sarnari F, di Michele A, Pascucci L, de Franco F, Zelante T, Antognelli C, Cruciani G, Talesa VN, Romani R, Fallarino F. Amniotic fluid stem cell-derived extracellular vesicles educate type 2 conventional dendritic cells to rescue autoimmune disorders in a multiple sclerosis mouse model. J Extracell Vesicles 2024; 13:e12446. [PMID: 38844736 PMCID: PMC11156524 DOI: 10.1002/jev2.12446] [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: 09/24/2023] [Revised: 04/11/2024] [Accepted: 04/15/2024] [Indexed: 06/10/2024] Open
Abstract
Dendritic cells (DCs) are essential orchestrators of immune responses and represent potential targets for immunomodulation in autoimmune diseases. Human amniotic fluid secretome is abundant in immunoregulatory factors, with extracellular vesicles (EVs) being a significant component. However, the impact of these EVs on dendritic cells subsets remain unexplored. In this study, we investigated the interaction between highly purified dendritic cell subsets and EVs derived from amniotic fluid stem cell lines (HAFSC-EVs). Our results suggest that HAFSC-EVs are preferentially taken up by conventional dendritic cell type 2 (cDC2) through CD29 receptor-mediated internalization, resulting in a tolerogenic DC phenotype characterized by reduced expression and production of pro-inflammatory mediators. Furthermore, treatment of cDC2 cells with HAFSC-EVs in coculture systems resulted in a higher proportion of T cells expressing the regulatory T cell marker Foxp3 compared to vehicle-treated control cells. Moreover, transfer of HAFSC-EV-treated cDC2s into an EAE mouse model resulted in the suppression of autoimmune responses and clinical improvement. These results suggest that HAFSC-EVs may serve as a promising tool for reprogramming inflammatory cDC2s towards a tolerogenic phenotype and for controlling autoimmune responses in the central nervous system, representing a potential platform for the study of the effects of EVs in DC subsets.
Collapse
Affiliation(s)
- Giorgia Manni
- Department of Medicine and SurgeryUniversity of PerugiaPerugiaItaly
- Extracellular Vesicles network (EV‐net) of the University of PerugiaPerugiaItaly
| | - Marco Gargaro
- Department of Pharmaceutical ScienceUniversity of PerugiaPerugiaItaly
| | - Doriana Ricciuti
- Department of Medicine and SurgeryUniversity of PerugiaPerugiaItaly
| | - Simona Fontana
- Department of Biomedicine, Neurosciences and advanced Diagnostics (Bi.N.D) School of MedicineUniversity of PalermoPalermoItaly
| | | | | | - Tommaso Mazza
- Bioinformatics unit, Fondazione IRCCS Casa Sollievo della SofferenzaSan Giovanni RotondoItaly
| | - Jessica Rosati
- Cellular Reprogramming Unit, Fondazione IRCCS Casa Sollievo della SofferenzaSan Giovanni RotondoItaly
| | - Alessandra di Veroli
- Department of Chemistry, Biology and BiotechnologyUniversity of PerugiaPerugiaItaly
| | | | | | | | - Giulia Scalisi
- Department of Medicine and SurgeryUniversity of PerugiaPerugiaItaly
| | | | - Alessandro di Michele
- Extracellular Vesicles network (EV‐net) of the University of PerugiaPerugiaItaly
- Department of Physics and GeologyUniversity of PerugiaPerugiaItaly
| | - Luisa Pascucci
- Extracellular Vesicles network (EV‐net) of the University of PerugiaPerugiaItaly
- Department of Veterinary MedicineUniversity of PerugiaPerugiaItaly
| | | | - Teresa Zelante
- Department of Medicine and SurgeryUniversity of PerugiaPerugiaItaly
| | | | - Gabriele Cruciani
- Department of Chemistry, Biology and BiotechnologyUniversity of PerugiaPerugiaItaly
| | | | - Rita Romani
- Department of Medicine and SurgeryUniversity of PerugiaPerugiaItaly
- Extracellular Vesicles network (EV‐net) of the University of PerugiaPerugiaItaly
| | - Francesca Fallarino
- Department of Medicine and SurgeryUniversity of PerugiaPerugiaItaly
- Extracellular Vesicles network (EV‐net) of the University of PerugiaPerugiaItaly
| |
Collapse
|
1073
|
Abyadeh M, Mirshahvaladi S, Kashani SA, Paulo JA, Amirkhani A, Mehryab F, Seydi H, Moradpour N, Jodeiryjabarzade S, Mirzaei M, Gupta V, Shekari F, Salekdeh GH. Proteomic profiling of mesenchymal stem cell-derived extracellular vesicles: Impact of isolation methods on protein cargo. JOURNAL OF EXTRACELLULAR BIOLOGY 2024; 3:e159. [PMID: 38947171 PMCID: PMC11212298 DOI: 10.1002/jex2.159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 03/01/2024] [Accepted: 05/15/2024] [Indexed: 07/02/2024]
Abstract
Extracellular vesicles (EVs) are nanosized vesicles with a lipid bilayer that are secreted by cells and play a critical role in cell-to-cell communication. Despite the promising reports regarding their diagnostic and therapeutic potential, the utilization of EVs in the clinical setting is limited due to insufficient information about their cargo and a lack of standardization in isolation and analysis methods. Considering protein cargos in EVs as key contributors to their therapeutic potency, we conducted a tandem mass tag (TMT) quantitative proteomics analysis of three subpopulations of mesenchymal stem cell (MSC)-derived EVs obtained through three different isolation techniques: ultracentrifugation (UC), high-speed centrifugation (HS), and ultracentrifugation on sucrose cushion (SU). Subsequently, we checked EV marker expression, size distribution, and morphological characterization, followed by bioinformatic analysis. The bioinformatic analysis of the proteome results revealed that these subpopulations exhibit distinct molecular and functional characteristics. The choice of isolation method impacts the proteome of isolated EVs by isolating different subpopulations of EVs. Specifically, EVs isolated through the high-speed centrifugation (HS) method exhibited a higher abundance of ribosomal and mitochondrial proteins. Functional apoptosis assays comparing isolated mitochondria with EVs isolated through different methods revealed that HS-EVs, but not other EVs, induced early apoptosis in cancer cells. On the other hand, EVs isolated using the sucrose cushion (SU) and ultracentrifugation (UC) methods demonstrated a higher abundance of proteins primarily involved in the immune response, cell-cell interactions and extracellular matrix interactions. Our analyses unveil notable disparities in proteins and associated biological functions among EV subpopulations, underscoring the importance of meticulously selecting isolation methods and resultant EV subpopulations based on the intended application.
Collapse
Affiliation(s)
- Morteza Abyadeh
- Department of Stem Cells and Developmental Biology, Cell Science Research CenterRoyan Institute for Stem Cell Biology and Technology, ACECRTehranIran
| | - Shahab Mirshahvaladi
- Macquarie Medical School, School of MedicineHealth and Human Sciences, Macquarie UniversitySydneyNew South WalesAustralia
| | - Sara Assar Kashani
- Department of Stem Cells and Developmental Biology, Cell Science Research CenterRoyan Institute for Stem Cell Biology and Technology, ACECRTehranIran
- Motor Neuron Disease Research Centre, Faculty of Medicine, Health and Human SciencesMacquarie UniversitySydneyNew South WalesAustralia
| | - Joao A. Paulo
- Department of Cell BiologyHarvard Medical SchoolBostonMassachusettsUSA
| | - Ardeshir Amirkhani
- Australian Proteome Analysis FacilityMacquarie UniversitySydneyNew South WalesAustralia
| | - Fatemeh Mehryab
- Advanced Therapy Medicinal Product Technology Development Center, Cell Science Research CenterRoyan Institute for Stem Cell Biology and Technology, ACECRTehranIran
| | - Homeyra Seydi
- Advanced Therapy Medicinal Product Technology Development Center, Cell Science Research CenterRoyan Institute for Stem Cell Biology and Technology, ACECRTehranIran
- Department of BiologyUniversity of Science and CultureTehranIran
| | | | | | - Mehdi Mirzaei
- Macquarie Medical School, School of MedicineHealth and Human Sciences, Macquarie UniversitySydneyNew South WalesAustralia
| | - Vivek Gupta
- Macquarie Medical School, School of MedicineHealth and Human Sciences, Macquarie UniversitySydneyNew South WalesAustralia
| | - Faezeh Shekari
- Department of Stem Cells and Developmental Biology, Cell Science Research CenterRoyan Institute for Stem Cell Biology and Technology, ACECRTehranIran
- Advanced Therapy Medicinal Product Technology Development Center, Cell Science Research CenterRoyan Institute for Stem Cell Biology and Technology, ACECRTehranIran
| | | |
Collapse
|
1074
|
Padinharayil H, George A. Small extracellular vesicles: Multi-functional aspects in non-small cell lung carcinoma. Crit Rev Oncol Hematol 2024; 198:104341. [PMID: 38575042 DOI: 10.1016/j.critrevonc.2024.104341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 03/13/2024] [Accepted: 03/28/2024] [Indexed: 04/06/2024] Open
Abstract
Extracellular vesicles (EVs) impact normal and pathological cellular signaling through bidirectional trafficking. Exosomes, a subset of EVs possess biomolecules including proteins, lipids, DNA fragments and various RNA species reflecting a speculum of their parent cells. The involvement of exosomes in bidirectional communication and their biological constituents substantiate its role in regulating both physiology and pathology, including multiple cancers. Non-small cell lung cancer (NSCLC) is the most common lung cancers (85%) with high incidence, mortality and reduced overall survival. Lack of efficient early diagnostic and therapeutic tools hurdles the management of NSCLC. Interestingly, the exosomes from body fluids similarity with parent cells or tissue offers a potential future multicomponent tool for the early diagnosis of NSCLC. The structural twinning of exosomes with a cell/tissue and the competitive tumor derived exosomes in tumor microenvironment (TME) promotes the unpinning horizons of exosomes as a drug delivery, vaccine, and therapeutic agent. Exosomes in clinical point of view assist to trace: acquired resistance caused by various therapeutic agents, early diagnosis, progression, and surveillance. In an integrated approach, EV biomarkers offer potential cutting-edge techniques for the detection and diagnosis of cancer, though the purification, characterization, and biomarker identification processes for the translational research regarding EVs need further optimization.
Collapse
Affiliation(s)
- Hafiza Padinharayil
- Jubilee Centre for Medical Research, Jubilee Mission Medical College and Research Institute, Thrissur-05, Kerala, India
| | - Alex George
- Jubilee Centre for Medical Research, Jubilee Mission Medical College and Research Institute, Thrissur-05, Kerala, India.
| |
Collapse
|
1075
|
Hansen DR, Svenningsen P. New guidelines to uncover the physiology of extracellular vesicles. Acta Physiol (Oxf) 2024; 240:e14153. [PMID: 38660772 DOI: 10.1111/apha.14153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 04/11/2024] [Accepted: 04/15/2024] [Indexed: 04/26/2024]
Affiliation(s)
- Didde R Hansen
- Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Per Svenningsen
- Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
1076
|
Afridi S, Sharma P, Choudhary F, Rizwan A, Nizam A, Parvez A, Farooqi H. Extracellular Vesicles: A New Approach to Study the Brain's Neural System and Its Diseases. Cell Biochem Biophys 2024; 82:521-534. [PMID: 38727784 DOI: 10.1007/s12013-024-01271-3] [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] [Accepted: 04/08/2024] [Indexed: 08/25/2024]
Abstract
In normal and pathophysiological conditions our cells secrete vesicular bodies known as extracellular particles. Extracellular vesicles are lipid-bound extracellular particles. A majority of these extracellular vesicles are linked to cell-to-cell communication. Brain consists of tightly packed neural cells. Neural cell releases extracellular vesicles in cerebrospinal fluid. Extracellular vesicle mediated crosstalk maintains neural homeostasis in the central nervous system via transferring cargos between neural cells. In neurodegenerative diseases, small extracellular vesicle transfer misfolded proteins to healthy cells in the neural microenvironment. They can also cross blood-brain barrier (BBB) and stimulate peripheral immune response inside central nervous system. In today's world different approaches employ extracellular vesicle in various therapeutics. This review gives a brief knowledge about the biological relevance of extracellular vesicles in the central nervous system and relevant advances in the translational application of EV in brain disorders.
Collapse
Affiliation(s)
- Shahid Afridi
- Department of Biotechnology, School of Chemical and Life Sciences, Jamia Hamdard, Hamdard Nagar, New Delhi, 110062, India
| | - Pradakshina Sharma
- Department of Biotechnology, School of Chemical and Life Sciences, Jamia Hamdard, Hamdard Nagar, New Delhi, 110062, India
| | - Furqan Choudhary
- Department of Biotechnology, School of Chemical and Life Sciences, Jamia Hamdard, Hamdard Nagar, New Delhi, 110062, India
| | - Amber Rizwan
- Department of Biotechnology, School of Chemical and Life Sciences, Jamia Hamdard, Hamdard Nagar, New Delhi, 110062, India
| | - Anam Nizam
- Department of Biotechnology, School of Chemical and Life Sciences, Jamia Hamdard, Hamdard Nagar, New Delhi, 110062, India
| | - Adil Parvez
- Department of Biotechnology, School of Chemical and Life Sciences, Jamia Hamdard, Hamdard Nagar, New Delhi, 110062, India
| | - Humaira Farooqi
- Department of Biotechnology, School of Chemical and Life Sciences, Jamia Hamdard, Hamdard Nagar, New Delhi, 110062, India.
| |
Collapse
|
1077
|
Gordillo-Sampedro S, Antounians L, Wei W, Mufteev M, Lendemeijer B, Kushner SA, de Vrij FMS, Zani A, Ellis J. iPSC-derived healthy human astrocytes selectively load miRNAs targeting neuronal genes into extracellular vesicles. Mol Cell Neurosci 2024; 129:103933. [PMID: 38663691 DOI: 10.1016/j.mcn.2024.103933] [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: 01/15/2024] [Revised: 03/31/2024] [Accepted: 04/20/2024] [Indexed: 05/05/2024] Open
Abstract
Astrocytes are in constant communication with neurons during the establishment and maturation of functional networks in the developing brain. Astrocytes release extracellular vesicles (EVs) containing microRNA (miRNA) cargo that regulates transcript stability in recipient cells. Astrocyte released factors are thought to be involved in neurodevelopmental disorders. Healthy astrocytes partially rescue Rett Syndrome (RTT) neuron function. EVs isolated from stem cell progeny also correct aspects of RTT. EVs cross the blood-brain barrier (BBB) and their cargo is found in peripheral blood which may allow non-invasive detection of EV cargo as biomarkers produced by healthy astrocytes. Here we characterize miRNA cargo and sequence motifs in healthy human astrocyte derived EVs (ADEVs). First, human induced Pluripotent Stem Cells (iPSC) were differentiated into Neural Progenitor Cells (NPCs) and subsequently into astrocytes using a rapid differentiation protocol. iPSC derived astrocytes expressed specific markers, displayed intracellular calcium transients and secreted ADEVs. miRNAs were identified by RNA-Seq on astrocytes and ADEVs and target gene pathway analysis detected brain and immune related terms. The miRNA profile was consistent with astrocyte identity, and included approximately 80 miRNAs found in astrocytes that were relatively depleted in ADEVs suggestive of passive loading. About 120 miRNAs were relatively enriched in ADEVs and motif analysis discovered binding sites for RNA binding proteins FUS, SRSF7 and CELF5. miR-483-5p was the most significantly enriched in ADEVs. This miRNA regulates MECP2 expression in neurons and has been found differentially expressed in blood samples from RTT patients. Our results identify potential miRNA biomarkers selectively sorted into ADEVs and implicate RNA binding protein sequence dependent mechanisms for miRNA cargo loading.
Collapse
Affiliation(s)
- Sara Gordillo-Sampedro
- Program in Developmental and Stem Cell Biology, Hospital for Sick Children, Toronto, ON, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Lina Antounians
- Program in Developmental and Stem Cell Biology, Hospital for Sick Children, Toronto, ON, Canada; Division of General and Thoracic Surgery, Hospital for Sick Children, Toronto, ON, Canada
| | - Wei Wei
- Program in Developmental and Stem Cell Biology, Hospital for Sick Children, Toronto, ON, Canada
| | - Marat Mufteev
- Program in Developmental and Stem Cell Biology, Hospital for Sick Children, Toronto, ON, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Bas Lendemeijer
- Department of Psychiatry, Erasmus University Medical Center, Rotterdam, The Netherlands; Department of Psychiatry, Columbia University Medical Center, New York, NY, USA
| | - Steven A Kushner
- Department of Psychiatry, Erasmus University Medical Center, Rotterdam, The Netherlands; Department of Psychiatry, Columbia University Medical Center, New York, NY, USA
| | - Femke M S de Vrij
- Department of Psychiatry, Erasmus University Medical Center, Rotterdam, The Netherlands; Center of Expertise for Neurodevelopmental Disorders (ENCORE), Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Augusto Zani
- Program in Developmental and Stem Cell Biology, Hospital for Sick Children, Toronto, ON, Canada; Division of General and Thoracic Surgery, Hospital for Sick Children, Toronto, ON, Canada; Department of Surgery, University of Toronto, Toronto, ON, Canada
| | - James Ellis
- Program in Developmental and Stem Cell Biology, Hospital for Sick Children, Toronto, ON, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
1078
|
Vaiaki EM, Falasca M. Comparative analysis of the minimal information for studies of extracellular vesicles guidelines: Advancements and implications for extracellular vesicle research. Semin Cancer Biol 2024; 101:12-24. [PMID: 38657746 DOI: 10.1016/j.semcancer.2024.04.002] [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: 02/28/2024] [Revised: 04/10/2024] [Accepted: 04/18/2024] [Indexed: 04/26/2024]
Abstract
In 2014, the International Society for Extracellular Vesicles (ISEV) introduced the Minimal Information for Studies of Extracellular Vesicles (MISEV) guidelines to establish standards for extracellular vesicle (EV) research. These guidelines aimed to enhance reliability and reproducibility, addressing the expanding field of EV science. EVs, membrane-bound particles released by cells, play crucial roles in intercellular communication and are potential biomarkers for various conditions. Over the years, the EV landscape witnessed a surge in publications, emphasizing their roles in cancer and immune modulation. In response, the MISEV guidelines underwent evolution, leading to the MISEV2018 update. This version, generated through community outreach, provided a comprehensive framework for EV research methodologies, emphasizing separation, characterization, reporting standards, and community engagement. The MISEV2018 guidelines reflected responsiveness to feedback, acknowledging the evolving EV research landscape. The guidelines served as a testament to the commitment of the scientific community to rigorous standards and the collective discernment of experts. The present article compares previous MISEV guidelines with its 2023 counterpart, highlighting advancements, changes, and impacts on EV research standardization. The 2023 guidelines build upon the 2018 principles, offering new recommendations for emerging areas. This comparative exploration contributes to understanding the transformative journey in EV research, emphasizing MISEV's pivotal role and the scientific community's adaptability to challenges.
Collapse
Affiliation(s)
- Elisavet Maria Vaiaki
- University of Parma, Department of Medicine and Surgery, Via Volturno 39, Parma 43125, Italy
| | - Marco Falasca
- University of Parma, Department of Medicine and Surgery, Via Volturno 39, Parma 43125, Italy.
| |
Collapse
|
1079
|
Jing R, Zhang L, Li R, Yang Z, Song J, Wang Q, Cao N, Han G, Yin H. Milk-derived extracellular vesicles functionalized with anti-tumour necrosis factor-α nanobody and anti-microbial peptide alleviate ulcerative colitis in mice. J Extracell Vesicles 2024; 13:e12462. [PMID: 38840457 PMCID: PMC11154809 DOI: 10.1002/jev2.12462] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 04/16/2024] [Accepted: 05/22/2024] [Indexed: 06/07/2024] Open
Abstract
Ulcerative colitis (UC) manifests clinically with chronic intestinal inflammation and microflora dysbiosis. Although biologics can effectively control inflammation, efficient delivery to the colon and colon epithelial cells remains challenging. Milk-derived extracellular vesicles (EV) show promise as an oral delivery tool, however, the ability to load biologics into EV presents challenges to therapeutic applications. Here, we demonstrate that fusing cell-penetrating peptide (TAT) to green fluorescent protein (GFP) enabled biologics loading into EV and protected against degradation in the gastrointestinal environment in vitro and in vivo after oral delivery. Oral administration of EV loaded with anti-tumour necrosis factor-α (TNF-α) nanobody (VHHm3F) (EVVHH) via TAT significantly reduced tissue TNF-α levels and alleviated pathologies in mice with acute UC, compared to VHH alone. In mice with chronic UC, simultaneously introducing VHH and an antimicrobial peptide LL37 into EV (EVLV), then administering orally improved intestinal barrier, inflammation and microbiota balance, resulted in relief of UC-induced depression and anxiety. Collectively, we demonstrated that oral delivery of EVLV effectively alleviated UC in mice and TAT efficiently loaded biologics into EV to confer protection from degradation in the gastrointestinal tract. This therapeutic strategy is promising for UC and is a simple and generalizable approach towards drug-loaded orally-administrable EV treatment for other diseases.
Collapse
Affiliation(s)
- Renwei Jing
- State Key Laboratory of Experimental Hematology & The Province and Ministry Co‐sponsored Collaborative Innovation Center for Medical Epigenetics & Key Laboratory of Immune Microenvironment and Disease (Ministry of Education) & School of Basic Medical Sciences & School of Medical TechnologyTianjin Medical UniversityTianjinChina
| | - Leijie Zhang
- State Key Laboratory of Experimental Hematology & The Province and Ministry Co‐sponsored Collaborative Innovation Center for Medical Epigenetics & Key Laboratory of Immune Microenvironment and Disease (Ministry of Education) & School of Basic Medical Sciences & School of Medical TechnologyTianjin Medical UniversityTianjinChina
| | - Ruibin Li
- State Key Laboratory of Experimental Hematology & The Province and Ministry Co‐sponsored Collaborative Innovation Center for Medical Epigenetics & Key Laboratory of Immune Microenvironment and Disease (Ministry of Education) & School of Basic Medical Sciences & School of Medical TechnologyTianjin Medical UniversityTianjinChina
| | - Zhongqiu Yang
- State Key Laboratory of Experimental Hematology & The Province and Ministry Co‐sponsored Collaborative Innovation Center for Medical Epigenetics & Key Laboratory of Immune Microenvironment and Disease (Ministry of Education) & School of Basic Medical Sciences & School of Medical TechnologyTianjin Medical UniversityTianjinChina
| | - Jun Song
- State Key Laboratory of Experimental Hematology & The Province and Ministry Co‐sponsored Collaborative Innovation Center for Medical Epigenetics & Key Laboratory of Immune Microenvironment and Disease (Ministry of Education) & School of Basic Medical Sciences & School of Medical TechnologyTianjin Medical UniversityTianjinChina
| | - Qian Wang
- State Key Laboratory of Experimental Hematology & The Province and Ministry Co‐sponsored Collaborative Innovation Center for Medical Epigenetics & Key Laboratory of Immune Microenvironment and Disease (Ministry of Education) & School of Basic Medical Sciences & School of Medical TechnologyTianjin Medical UniversityTianjinChina
| | - Nan Cao
- State Key Laboratory of Experimental Hematology & The Province and Ministry Co‐sponsored Collaborative Innovation Center for Medical Epigenetics & Key Laboratory of Immune Microenvironment and Disease (Ministry of Education) & School of Basic Medical Sciences & School of Medical TechnologyTianjin Medical UniversityTianjinChina
| | - Gang Han
- State Key Laboratory of Experimental Hematology & The Province and Ministry Co‐sponsored Collaborative Innovation Center for Medical Epigenetics & Key Laboratory of Immune Microenvironment and Disease (Ministry of Education) & School of Basic Medical Sciences & School of Medical TechnologyTianjin Medical UniversityTianjinChina
| | - HaiFang Yin
- State Key Laboratory of Experimental Hematology & The Province and Ministry Co‐sponsored Collaborative Innovation Center for Medical Epigenetics & Key Laboratory of Immune Microenvironment and Disease (Ministry of Education) & School of Basic Medical Sciences & School of Medical TechnologyTianjin Medical UniversityTianjinChina
- Department of Clinical LaboratoryTianjin Medical University General HospitalTianjinChina
| |
Collapse
|
1080
|
Nadeau A, Burnier JV. With >1000 authors, the updated "Minimum Information for Studies of Extracellular Vesicles" (MISEV) promotes rigor, reproducibility, and transparency in the liquid biopsy field. THE JOURNAL OF LIQUID BIOPSY 2024; 4:100147. [PMID: 40027143 PMCID: PMC11863875 DOI: 10.1016/j.jlb.2024.100147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 02/08/2024] [Indexed: 03/05/2025]
Affiliation(s)
- Amélie Nadeau
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- Department of Pathology, McGill University, Montreal, QC, Canada
| | - Julia V Burnier
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- Department of Pathology, McGill University, Montreal, QC, Canada
- Gerald Bronfman Department of Oncology, McGill University, Montreal, QC, Canada
| |
Collapse
|
1081
|
Grätz C, Schuster M, Brandes F, Meidert AS, Kirchner B, Reithmair M, Schelling G, Pfaffl MW. A pipeline for the development and analysis of extracellular vesicle-based transcriptomic biomarkers in molecular diagnostics. Mol Aspects Med 2024; 97:101269. [PMID: 38552453 DOI: 10.1016/j.mam.2024.101269] [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: 12/01/2023] [Revised: 03/11/2024] [Accepted: 03/17/2024] [Indexed: 06/12/2024]
Abstract
Extracellular vesicles are shed by every cell type and can be found in any biofluid. They contain different molecules that can be utilized as biomarkers, including several RNA species which they protect from degradation. Here, we present a pipeline for the development and analysis of extracellular vesicle-associated transcriptomic biomarkers that our group has successfully applied multiple times. We highlight the key steps of the pipeline and give particular emphasis to the necessary quality control checkpoints, which are linked to numerous available guidelines that should be considered along the workflow. Our pipeline starts with patient recruitment and continues with blood sampling and processing. The purification and characterization of extracellular vesicles is explained in detail, as well as the isolation and quality control of extracellular vesicle-associated RNA. We point out the possible pitfalls during library preparation and RNA sequencing and present multiple bioinformatic tools to pinpoint biomarker signature candidates from the sequencing data. Finally, considerations and pitfalls during the validation of the biomarker signature using RT-qPCR will be elaborated.
Collapse
Affiliation(s)
- Christian Grätz
- Department of Animal Physiology and Immunology, School of Life Sciences, Technical University of Munich, Freising, Germany.
| | - Martina Schuster
- Institute of Human Genetics, University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Florian Brandes
- Department of Anesthesiology, University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Agnes S Meidert
- Department of Anesthesiology, University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Benedikt Kirchner
- Department of Animal Physiology and Immunology, School of Life Sciences, Technical University of Munich, Freising, Germany; Institute of Human Genetics, University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Marlene Reithmair
- Institute of Human Genetics, University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Gustav Schelling
- Department of Anesthesiology, University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Michael W Pfaffl
- Department of Animal Physiology and Immunology, School of Life Sciences, Technical University of Munich, Freising, Germany.
| |
Collapse
|
1082
|
Ma W, Wu D, Long C, Liu J, Xu L, Zhou L, Dou Q, Ge Y, Zhou C, Jia R. Neutrophil-derived nanovesicles deliver IL-37 to mitigate renal ischemia-reperfusion injury via endothelial cell targeting. J Control Release 2024; 370:66-81. [PMID: 38631490 DOI: 10.1016/j.jconrel.2024.04.025] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 04/09/2024] [Accepted: 04/14/2024] [Indexed: 04/19/2024]
Abstract
Renal ischemia-reperfusion injury (IRI) is one of the most important causes of acute kidney injury (AKI). Interleukin (IL)-37 has been suggested as a novel anti-inflammatory factor for the treatment of IRI, but its application is still limited by its low stability and delivery efficiency. In this study, we reported a novel engineered method to efficiently and easily prepare neutrophil membrane-derived vesicles (N-MVs), which could be utilized as a promising vehicle to deliver IL-37 and avoid the potential side effects of neutrophil-derived natural extracellular vesicles. N-MVs could enhance the stability of IL-37 and targetedly deliver IL-37 to damaged endothelial cells of IRI kidneys via P-selectin glycoprotein ligand-1 (PSGL-1). In vitro and in vivo evidence revealed that N-MVs encapsulated with IL-37 (N-MV@IL-37) could inhibit endothelial cell apoptosis, promote endothelial cell proliferation and angiogenesis, and decrease inflammatory factor production and leukocyte infiltration, thereby ameliorating renal IRI. Our study establishes a promising delivery vehicle for the treatment of renal IRI and other endothelial damage-related diseases.
Collapse
Affiliation(s)
- Wenjie Ma
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing 210006, China
| | - Di Wu
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing 210006, China
| | - Chengcheng Long
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing 210006, China
| | - Jingyu Liu
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing 210006, China
| | - Luwei Xu
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing 210006, China
| | - Liuhua Zhou
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing 210006, China
| | - Quanliang Dou
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing 210006, China
| | - Yuzheng Ge
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing 210006, China
| | - Changcheng Zhou
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing 210006, China.
| | - Ruipeng Jia
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing 210006, China.
| |
Collapse
|
1083
|
Klangprapan J, Souza GR, Ferreira JN. Bioprinting salivary gland models and their regenerative applications. BDJ Open 2024; 10:39. [PMID: 38816372 PMCID: PMC11139920 DOI: 10.1038/s41405-024-00219-2] [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: 01/12/2024] [Revised: 04/29/2024] [Accepted: 04/30/2024] [Indexed: 06/01/2024] Open
Abstract
OBJECTIVE Salivary gland (SG) hypofunction is a common clinical condition arising from radiotherapy to suppress head and neck cancers. The radiation often destroys the SG secretory acini, and glands are left with limited regenerative potential. Due to the complex architecture of SG acini and ducts, three-dimensional (3D) bioprinting platforms have emerged to spatially define these in vitro epithelial units and develop mini-organs or organoids for regeneration. Due to the limited body of evidence, this comprehensive review highlights the advantages and challenges of bioprinting platforms for SG regeneration. METHODS SG microtissue engineering strategies such as magnetic 3D bioassembly of cells and microfluidic coaxial 3D bioprinting of cell-laden microfibers and microtubes have been proposed to replace the damaged acinar units, avoid the use of xenogeneic matrices (like Matrigel), and restore salivary flow. RESULTS Replacing the SG damaged organ is challenging due to its complex architecture, which combines a ductal network with acinar epithelial units to facilitate a unidirectional flow of saliva. Our research group was the first to develop 3D bioassembly SG epithelial functional organoids with innervation to respond to both cholinergic and adrenergic stimulation. More recently, microtissue engineering using coaxial 3D bioprinting of hydrogel microfibers and microtubes could also supported the formation of viable epithelial units. Both bioprinting approaches could overcome the need for Matrigel by facilitating the assembly of adult stem cells, such as human dental pulp stem cells, and primary SG cells into micro-sized 3D constructs able to produce their own matrix and self-organize into micro-modular tissue clusters with lumenized areas. Furthermore, extracellular vesicle (EV) therapies from organoid-derived secretome were also designed and validated ex vivo for SG regeneration after radiation damage. CONCLUSION Magnetic 3D bioassembly and microfluidic coaxial bioprinting platforms have the potential to create SG mini-organs for regenerative applications via organoid transplantation or organoid-derived EV therapies.
Collapse
Affiliation(s)
- Jutapak Klangprapan
- Avatar Biotechnologies for Oral Health and Healthy Longevity Research Unit, Faculty of Dentistry, Chulalongkorn University, 34 Henri-Dunant Road, Pathumwan, Bangkok, 10330, Thailand
| | - Glauco R Souza
- Greiner Bio-one North America Inc., 4238 Capital Drive, Monroe, NC, 28110, USA
| | - João N Ferreira
- Avatar Biotechnologies for Oral Health and Healthy Longevity Research Unit, Faculty of Dentistry, Chulalongkorn University, 34 Henri-Dunant Road, Pathumwan, Bangkok, 10330, Thailand.
| |
Collapse
|
1084
|
Leone I, Santoro J, Soricelli A, Febbraro A, Santoriello A, Carrese B. Triple-Negative Breast Cancer EVs Modulate Growth and Migration of Normal Epithelial Lung Cells. Int J Mol Sci 2024; 25:5864. [PMID: 38892050 PMCID: PMC11172765 DOI: 10.3390/ijms25115864] [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: 04/24/2024] [Revised: 05/20/2024] [Accepted: 05/23/2024] [Indexed: 06/21/2024] Open
Abstract
Breast cancer is the most common cancer amongst women worldwide. Recently, owing to screening programs and new technologies, the survival rate has increased significantly. Breast cancer can potentially develop metastases, and, despite them, lung metastases generally occur within five years of breast cancer diagnosis. In this study, the objective was to analyze the effect of breast cancer-derived EVs on a lung epithelial cell line. BEAS-2B cells were treated with extracellular vesicles (EVs) derived from triple-negative breast cancer cells (TNBCs), e.g., MDA-MB-231 and HS578T, separated using differential ultracentrifugation. We observed an increased growth, migration, and invasiveness of normal epithelial lung cells over time in the presence of TNBC EVs compared to the control. Therefore, these data suggest that EVs released by tumor cells contain biological molecules capable of influencing the pro-tumorigenic activity of normal cells. Exploring the role of EVs in oncology research and their potential cargo may be novel biomarkers for early cancer detection and further diagnosis.
Collapse
Affiliation(s)
- Ilaria Leone
- IRCCS SYNLAB SDN, Via E. Gianturco, 80143 Naples, Italy; (I.L.); (A.S.); (B.C.)
| | - Jessie Santoro
- IRCCS SYNLAB SDN, Via E. Gianturco, 80143 Naples, Italy; (I.L.); (A.S.); (B.C.)
| | - Andrea Soricelli
- IRCCS SYNLAB SDN, Via E. Gianturco, 80143 Naples, Italy; (I.L.); (A.S.); (B.C.)
| | - Antonio Febbraro
- Oncology Unit, Casa di Cura Cobellis, Vallo della Lucania, 84078 Vallo della Lucania, Italy;
| | - Antonio Santoriello
- Breast Unit, Casa di Cura Cobellis, Vallo della Lucania, 84078 Vallo della Lucania, Italy;
| | - Barbara Carrese
- IRCCS SYNLAB SDN, Via E. Gianturco, 80143 Naples, Italy; (I.L.); (A.S.); (B.C.)
| |
Collapse
|
1085
|
Yavuz B, Mutlu EC, Ahmed Z, Ben-Nissan B, Stamboulis A. Applications of Stem Cell-Derived Extracellular Vesicles in Nerve Regeneration. Int J Mol Sci 2024; 25:5863. [PMID: 38892052 PMCID: PMC11172915 DOI: 10.3390/ijms25115863] [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: 03/06/2024] [Revised: 05/15/2024] [Accepted: 05/23/2024] [Indexed: 06/21/2024] Open
Abstract
Extracellular vesicles (EVs), including exosomes, microvesicles, and other lipid vesicles derived from cells, play a pivotal role in intercellular communication by transferring information between cells. EVs secreted by progenitor and stem cells have been associated with the therapeutic effects observed in cell-based therapies, and they also contribute to tissue regeneration following injury, such as in orthopaedic surgery cases. This review explores the involvement of EVs in nerve regeneration, their potential as drug carriers, and their significance in stem cell research and cell-free therapies. It underscores the importance of bioengineers comprehending and manipulating EV activity to optimize the efficacy of tissue engineering and regenerative therapies.
Collapse
Affiliation(s)
- Burcak Yavuz
- Vocational School of Health Services, Altinbas University, 34147 Istanbul, Turkey;
| | - Esra Cansever Mutlu
- Biomaterials Research Group, School of Metallurgy and Materials, College of Engineering and Physical Science, University of Birmingham, Birmingham B15 2TT, UK;
| | - Zubair Ahmed
- Neuroscience & Ophthalmology, Institute of Inflammation and Ageing, University of Birmingham, Edgbaston B15 2TT, UK
| | - Besim Ben-Nissan
- Translational Biomaterials and Medicine Group, School of Life Sciences, University of Technology Sydney, P.O. Box 123, Broadway, NSW 2007, Australia;
| | - Artemis Stamboulis
- Biomaterials Research Group, School of Metallurgy and Materials, College of Engineering and Physical Science, University of Birmingham, Birmingham B15 2TT, UK;
| |
Collapse
|
1086
|
Yamada R, Michimae M, Hamamoto A, Takemori H. Melanin-concentrating hormone receptor 1 is discarded by exosomes after internalization. Biochem Biophys Res Commun 2024; 710:149917. [PMID: 38604071 DOI: 10.1016/j.bbrc.2024.149917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 04/01/2024] [Accepted: 04/06/2024] [Indexed: 04/13/2024]
Abstract
Melanin-concentrating hormone (MCH) receptor 1 (MCHR1), a G protein-coupled receptor, is poised for interaction with its ligands on the plasma membrane. Analyses of MCHR1 knockout mice suggest that this receptor could be a therapeutic target for the treatment of appetite disorders, glucose metabolism, psychiatric disorders, and inflammation. Binding of MCH to MCHR1 initiates calcium signaling, which is subsequently attenuated through receptor internalization. However, the ultimate destiny of the receptor post-internalization remains unexplored. In this study, we report the extracellular secretion of MCHR1 via exosomes. The recruitment of MCHR1 to exosomes occurs subsequent to its internalization, which is induced by stimulation with the ligand MCH. Although a highly glycosylated form of MCHR1, potentially representing a mature form, is selectively recruited to exosomes, the MCHR1 transferred into other cells does not exhibit functionality. The truncation of MCHR1 at the C-terminus not only impairs its response to MCH but also hinders its recruitment to exosomes. These findings imply that functional MCHR1 could be secreted extracellularly via exosomes, a process that may represent a mechanism for the termination of intracellular MCHR1 signaling.
Collapse
Affiliation(s)
- Ryohei Yamada
- Department of Chemistry and Biomolecular Science, Faculty of Engineering, Gifu University, 1-1 Yanagido, Gifu, 501-1193, Japan
| | - Momoka Michimae
- Department of Chemistry and Biomolecular Science, Faculty of Engineering, Gifu University, 1-1 Yanagido, Gifu, 501-1193, Japan
| | - Akie Hamamoto
- Department of Chemistry and Biomolecular Science, Faculty of Engineering, Gifu University, 1-1 Yanagido, Gifu, 501-1193, Japan; Graduate School of Integrated Arts and Sciences, Hiroshima University, Hiroshima, 739-8521, Japan
| | - Hiroshi Takemori
- Department of Chemistry and Biomolecular Science, Faculty of Engineering, Gifu University, 1-1 Yanagido, Gifu, 501-1193, Japan.
| |
Collapse
|
1087
|
Porcu C, Dobrowolny G, Scicchitano BM. Exploring the Role of Extracellular Vesicles in Skeletal Muscle Regeneration. Int J Mol Sci 2024; 25:5811. [PMID: 38892005 PMCID: PMC11171935 DOI: 10.3390/ijms25115811] [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: 04/30/2024] [Revised: 05/21/2024] [Accepted: 05/24/2024] [Indexed: 06/21/2024] Open
Abstract
Skeletal muscle regeneration entails a multifaceted process marked by distinct phases, encompassing inflammation, regeneration, and remodeling. The coordination of these phases hinges upon precise intercellular communication orchestrated by diverse cell types and signaling molecules. Recent focus has turned towards extracellular vesicles (EVs), particularly small EVs, as pivotal mediators facilitating intercellular communication throughout muscle regeneration. Notably, injured muscle provokes the release of EVs originating from myofibers and various cell types, including mesenchymal stem cells, satellite cells, and immune cells such as M2 macrophages, which exhibit anti-inflammatory and promyogenic properties. EVs harbor a specific cargo comprising functional proteins, lipids, and nucleic acids, including microRNAs (miRNAs), which intricately regulate gene expression in target cells and activate downstream pathways crucial for skeletal muscle homeostasis and repair. Furthermore, EVs foster angiogenesis, muscle reinnervation, and extracellular matrix remodeling, thereby modulating the tissue microenvironment and promoting effective tissue regeneration. This review consolidates the current understanding on EVs released by cells and damaged tissues throughout various phases of muscle regeneration with a focus on EV cargo, providing new insights on potential therapeutic interventions to mitigate muscle-related pathologies.
Collapse
Affiliation(s)
- Cristiana Porcu
- DAHFMO-Unità di Istologia ed Embriologia Medica, Sapienza Università di Roma, 00161 Roma, Italy;
| | - Gabriella Dobrowolny
- DAHFMO-Unità di Istologia ed Embriologia Medica, Sapienza Università di Roma, 00161 Roma, Italy;
| | - Bianca Maria Scicchitano
- Sezione di Istologia ed Embriologia, Dipartimento di Scienze della Vita e Sanità Pubblica, Università Cattolica del Sacro Cuore, 00168 Roma, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Roma, Italy
| |
Collapse
|
1088
|
Wu Y, Cao Y, Chen L, Lai X, Zhang S, Wang S. Role of Exosomes in Cancer and Aptamer-Modified Exosomes as a Promising Platform for Cancer Targeted Therapy. Biol Proced Online 2024; 26:15. [PMID: 38802766 PMCID: PMC11129508 DOI: 10.1186/s12575-024-00245-2] [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: 04/14/2024] [Accepted: 05/16/2024] [Indexed: 05/29/2024] Open
Abstract
Exosomes are increasingly recognized as important mediators of intercellular communication in cancer biology. Exosomes can be derived from cancer cells as well as cellular components in tumor microenvironment. After secretion, the exosomes carrying a wide range of bioactive cargos can be ingested by local or distant recipient cells. The released cargos act through a variety of mechanisms to elicit multiple biological effects and impact most if not all hallmarks of cancer. Moreover, owing to their excellent biocompatibility and capability of being easily engineered or modified, exosomes are currently exploited as a promising platform for cancer targeted therapy. In this review, we first summarize the current knowledge of roles of exosomes in risk and etiology, initiation and progression of cancer, as well as their underlying molecular mechanisms. The aptamer-modified exosome as a promising platform for cancer targeted therapy is then briefly introduced. We also discuss the future directions for emerging roles of exosome in tumor biology and perspective of aptamer-modified exosomes in cancer therapy.
Collapse
Affiliation(s)
- Yating Wu
- Fujian Key Laboratory of Aptamers Technology, Affiliated Dongfang Hospital of School of Medicine, Xiamen University, Fuzhou, Fujian Province, P. R. China
- Department of Medical Oncology, Fuzhou General Clinical Medical School (the 900th Hospital), Fujian Medical University, Fujian Province, Fuzhou, P. R. China
| | - Yue Cao
- Department of Clinical Laboratory Medicine, Fuzhou General Clinical Medical School (the 900 th Hospital), Fujian Medical University, Fujian Province, Fuzhou, P. R. China
| | - Li Chen
- Fujian Key Laboratory of Aptamers Technology, Affiliated Dongfang Hospital of School of Medicine, Xiamen University, Fuzhou, Fujian Province, P. R. China
- Department of Clinical Laboratory Medicine, Fuzhou General Clinical Medical School (the 900 th Hospital), Fujian Medical University, Fujian Province, Fuzhou, P. R. China
| | - Xiaofeng Lai
- Fujian Key Laboratory of Aptamers Technology, Affiliated Dongfang Hospital of School of Medicine, Xiamen University, Fuzhou, Fujian Province, P. R. China
- Department of Clinical Laboratory Medicine, Fuzhou General Clinical Medical School (the 900 th Hospital), Fujian Medical University, Fujian Province, Fuzhou, P. R. China
| | - Shenghang Zhang
- Fujian Key Laboratory of Aptamers Technology, Affiliated Dongfang Hospital of School of Medicine, Xiamen University, Fuzhou, Fujian Province, P. R. China.
- Department of Clinical Laboratory Medicine, Fuzhou General Clinical Medical School (the 900 th Hospital), Fujian Medical University, Fujian Province, Fuzhou, P. R. China.
| | - Shuiliang Wang
- Fujian Key Laboratory of Aptamers Technology, Affiliated Dongfang Hospital of School of Medicine, Xiamen University, Fuzhou, Fujian Province, P. R. China.
- Department of Clinical Laboratory Medicine, Fuzhou General Clinical Medical School (the 900 th Hospital), Fujian Medical University, Fujian Province, Fuzhou, P. R. China.
| |
Collapse
|
1089
|
Puletic M, Velikic G, Maric DM, Supic G, Maric DL, Radovic N, Avramov S, Vojvodic D. Clinical Efficacy of Extracellular Vesicle Therapy in Periodontitis: Reduced Inflammation and Enhanced Regeneration. Int J Mol Sci 2024; 25:5753. [PMID: 38891939 PMCID: PMC11171522 DOI: 10.3390/ijms25115753] [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: 04/27/2024] [Revised: 05/12/2024] [Accepted: 05/21/2024] [Indexed: 06/21/2024] Open
Abstract
Periodontitis, a prevalent inflammatory condition, affects the supporting structures of teeth, leading to significant oral health challenges. Traditional treatments have primarily focused on mechanical debridement, antimicrobial therapy, and surgery, which often fail to restore lost periodontal structures. Emerging as a novel approach in regenerative medicine, extracellular vesicle (EV) therapy, including exosomes, leverages nano-sized vesicles known for facilitating intercellular communication and modulating physiological and pathological processes. This study is a proof-of-concept type that evaluates the clinical efficacy of EV therapy as a non-surgical treatment for stage I-III periodontitis, focusing on its anti-inflammatory and regenerative potential. The research involved seven patients undergoing the therapy, and seven healthy individuals. Clinical parameters, including the plaque index, bleeding on probing, probing depth, and attachment level, were assessed alongside cytokine levels in the gingival crevicular fluid. The study found significant improvements in clinical parameters, and a marked reduction in pro-inflammatory cytokines post-treatment, matching the levels of healthy subjects, underscoring the therapy's ability to not only attenuate inflammation and enhance tissue regeneration, but also highlighting its potential in restoring periodontal health. This investigation illuminates the promising role of EV therapy in periodontal treatment, advocating for a shift towards therapies that halt disease progression and promote structural and functional restoration of periodontal tissues.
Collapse
Affiliation(s)
- Miljan Puletic
- Faculty of Stomatology Pancevo, University Business Academy, 26101 Pancevo, Serbia; (M.P.); (D.M.M.); (N.R.); (S.A.)
| | - Gordana Velikic
- Department for Research and Development, Clinic Orto MD-Parks Hospital, 21000 Novi Sad, Serbia
- Hajim School of Engineering, University of Rochester, Rochester, NY 14627, USA
| | - Dusan M. Maric
- Faculty of Stomatology Pancevo, University Business Academy, 26101 Pancevo, Serbia; (M.P.); (D.M.M.); (N.R.); (S.A.)
- Department for Research and Development, Clinic Orto MD-Parks Hospital, 21000 Novi Sad, Serbia
| | - Gordana Supic
- Institute for Medical Research, Military Medical Academy, 11000 Belgrade, Serbia; (G.S.); (D.V.)
- Medical Faculty of Military Medical Academy, University of Defense, 11000 Belgrade, Serbia
| | - Dusica L. Maric
- Department of Anatomy, Faculty of Medicine, University of Novi Sad, 21000 Novi Sad, Serbia
| | - Nikola Radovic
- Faculty of Stomatology Pancevo, University Business Academy, 26101 Pancevo, Serbia; (M.P.); (D.M.M.); (N.R.); (S.A.)
| | - Stevan Avramov
- Faculty of Stomatology Pancevo, University Business Academy, 26101 Pancevo, Serbia; (M.P.); (D.M.M.); (N.R.); (S.A.)
- Institute for Biological Research “Sinisa Stankovic”, National Institute of the Republic of Serbia, University of Belgrade, 11060 Belgrade, Serbia
| | - Danilo Vojvodic
- Institute for Medical Research, Military Medical Academy, 11000 Belgrade, Serbia; (G.S.); (D.V.)
- Medical Faculty of Military Medical Academy, University of Defense, 11000 Belgrade, Serbia
| |
Collapse
|
1090
|
Piibor J, Waldmann A, Prasadani M, Kavak A, Andronowska A, Klein C, Kodithuwakku S, Fazeli A. Investigation of Uterine Fluid Extracellular Vesicles' Proteomic Profiles Provides Novel Diagnostic Biomarkers of Bovine Endometritis. Biomolecules 2024; 14:626. [PMID: 38927030 PMCID: PMC11202259 DOI: 10.3390/biom14060626] [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: 04/24/2024] [Revised: 05/22/2024] [Accepted: 05/23/2024] [Indexed: 06/28/2024] Open
Abstract
Cow uterine infections pose a challenge in dairy farming, resulting in reproductive disorders. Uterine fluid extracellular vesicles (UF-EVs) play a key role in cell-to-cell communication in the uterus, potentially holding the signs of aetiology for endometritis. We used mass spectrometry-based quantitative shotgun proteomics to compare UF-EV proteomic profiles in healthy cows (H), cows with subclinical (SE) or clinical endometritis (CLE) sampled at 28-35 days postpartum. Functional analysis was performed on embryo cultures with the exposure to different EV types. A total of 248 UF-EV proteins exhibited differential enrichment between the groups. Interestingly, in SE, EV protein signature suggests a slight suppression of inflammatory response compared to CLE-UF-EVs, clustering closer with healthy cows' profile. Furthermore, CLE-UF-EVs proteomic profile highlighted pathways associated with cell apoptosis and active inflammation aimed at pathogen elimination. In SE-UF-EVs, the regulation of normal physiological status was aberrant, showing cell damage and endometrial repair at the same time. Serine peptidase HtrA1 (HTRA1) emerged as a potential biomarker for SE. Supplementation of CLE- and SE-derived UF-EVs reduced the embryo developmental rates and quality. Therefore, further research is warranted to elucidate the precise aetiology of SE in cattle, and HTRA1 should be further explored as a potential diagnostic biomarker.
Collapse
Affiliation(s)
- Johanna Piibor
- Institute of Veterinary Medicine and Animal Sciences, Estonian University of Life Sciences, Kreutzwaldi 62, 51006 Tartu, Estonia; (J.P.); (A.W.); (M.P.); (A.K.); (S.K.)
| | - Andres Waldmann
- Institute of Veterinary Medicine and Animal Sciences, Estonian University of Life Sciences, Kreutzwaldi 62, 51006 Tartu, Estonia; (J.P.); (A.W.); (M.P.); (A.K.); (S.K.)
- Faculty of Veterinary Medicine, Latvia University of Life Sciences and Technologies, LV-3004 Jelgava, Latvia
| | - Madhusha Prasadani
- Institute of Veterinary Medicine and Animal Sciences, Estonian University of Life Sciences, Kreutzwaldi 62, 51006 Tartu, Estonia; (J.P.); (A.W.); (M.P.); (A.K.); (S.K.)
| | - Ants Kavak
- Institute of Veterinary Medicine and Animal Sciences, Estonian University of Life Sciences, Kreutzwaldi 62, 51006 Tartu, Estonia; (J.P.); (A.W.); (M.P.); (A.K.); (S.K.)
| | - Aneta Andronowska
- Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Juliana Tuwima 10, 10-748 Olsztyn, Poland;
| | - Claudia Klein
- Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Höltystr. 10, 31535 Neustadt, Germany;
| | - Suranga Kodithuwakku
- Institute of Veterinary Medicine and Animal Sciences, Estonian University of Life Sciences, Kreutzwaldi 62, 51006 Tartu, Estonia; (J.P.); (A.W.); (M.P.); (A.K.); (S.K.)
- Department of Animal Sciences, Faculty of Agriculture, University of Peradeniya, Peradeniya 20400, Sri Lanka
| | - Alireza Fazeli
- Institute of Veterinary Medicine and Animal Sciences, Estonian University of Life Sciences, Kreutzwaldi 62, 51006 Tartu, Estonia; (J.P.); (A.W.); (M.P.); (A.K.); (S.K.)
- Department of Pathophysiology, Institute of Biomedicine and Translational Medicine, University of Tartu, Ravila St. 14b, 50411 Tartu, Estonia
- Division of Clinical Medicine, School of Medicine & Population Health, University of Sheffield, Level 4, Jessop Wing, Tree Root Walk, Sheffield S10 2SF, UK
| |
Collapse
|
1091
|
Chen X, Yang N, Li B, Gao X, Wang Y, Wang Q, Liu X, Zhang Z, Zhang R. Visualization Analysis of Small Extracellular Vesicles in the Application of Bone-Related Diseases. Cells 2024; 13:904. [PMID: 38891036 PMCID: PMC11171653 DOI: 10.3390/cells13110904] [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: 04/11/2024] [Revised: 05/17/2024] [Accepted: 05/20/2024] [Indexed: 06/20/2024] Open
Abstract
Small extracellular vesicles were shown to have similar functional roles to their parent cells without the defect of potential tumorigenicity, which made them a great candidate for regenerative medicine. The last twenty years have witnessed the rapid development of research on small extracellular vesicles. In this paper, we employed a scientometric synthesis method to conduct a retrospective analysis of small extracellular vesicles in the field of bone-related diseases. The overall background analysis consisted the visualization of the countries, institutions, journals, and authors involved in research. The current status of the research direction and future trends were presented through the analysis of references and keywords, which showed that engineering strategies, mesenchymal stem cell derived exosomes, and cartilage damage were the most concerning topics, and scaffold, osteoarthritis, platelet-rich plasma, and senescence were the future trends. We also discussed the current problems and challenges in practical applications, including the in-sight mechanisms, the building of relevant animal models, and the problems in clinical trials. By using CiteSpace, VOSviewer, and Bibliometrix, the presented data avoided subjective selectivity and tendency well, which made the conclusion more reliable and comprehensive. We hope that the findings can provide new perspectives for researchers to understand the evolution of this field over time and to search for novel research directions.
Collapse
Affiliation(s)
- Xinjiani Chen
- Department of Biotechnology and Biomedicine, Yangtze Delta Region Institute of Tsinghua University, Jiaxing 314006, China; (X.C.); (N.Y.); (B.L.); (X.G.); (Y.W.); (Q.W.); (X.L.)
- Ministry of Education Key Laboratory of Protein Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Ning Yang
- Department of Biotechnology and Biomedicine, Yangtze Delta Region Institute of Tsinghua University, Jiaxing 314006, China; (X.C.); (N.Y.); (B.L.); (X.G.); (Y.W.); (Q.W.); (X.L.)
| | - Bailei Li
- Department of Biotechnology and Biomedicine, Yangtze Delta Region Institute of Tsinghua University, Jiaxing 314006, China; (X.C.); (N.Y.); (B.L.); (X.G.); (Y.W.); (Q.W.); (X.L.)
- Ministry of Education Key Laboratory of Protein Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Xinyu Gao
- Department of Biotechnology and Biomedicine, Yangtze Delta Region Institute of Tsinghua University, Jiaxing 314006, China; (X.C.); (N.Y.); (B.L.); (X.G.); (Y.W.); (Q.W.); (X.L.)
| | - Yayu Wang
- Department of Biotechnology and Biomedicine, Yangtze Delta Region Institute of Tsinghua University, Jiaxing 314006, China; (X.C.); (N.Y.); (B.L.); (X.G.); (Y.W.); (Q.W.); (X.L.)
- Ministry of Education Key Laboratory of Protein Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Qin Wang
- Department of Biotechnology and Biomedicine, Yangtze Delta Region Institute of Tsinghua University, Jiaxing 314006, China; (X.C.); (N.Y.); (B.L.); (X.G.); (Y.W.); (Q.W.); (X.L.)
- Ministry of Education Key Laboratory of Protein Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Xiaojun Liu
- Department of Biotechnology and Biomedicine, Yangtze Delta Region Institute of Tsinghua University, Jiaxing 314006, China; (X.C.); (N.Y.); (B.L.); (X.G.); (Y.W.); (Q.W.); (X.L.)
- Zhejiang Provincial Key Laboratory of Applied Enzymology, Yangtze Delta Region Institute of Tsinghua University, 705 Yatai Road, Jiaxing 314006, China
- Taizhou Innovation Center, Yangtze Delta Region Institute of Tsinghua University, Jiaxing 318000, China
| | - Zhen Zhang
- Department of Biotechnology and Biomedicine, Yangtze Delta Region Institute of Tsinghua University, Jiaxing 314006, China; (X.C.); (N.Y.); (B.L.); (X.G.); (Y.W.); (Q.W.); (X.L.)
- Zhejiang Provincial Key Laboratory of Applied Enzymology, Yangtze Delta Region Institute of Tsinghua University, 705 Yatai Road, Jiaxing 314006, China
| | - Rongqing Zhang
- Department of Biotechnology and Biomedicine, Yangtze Delta Region Institute of Tsinghua University, Jiaxing 314006, China; (X.C.); (N.Y.); (B.L.); (X.G.); (Y.W.); (Q.W.); (X.L.)
- Ministry of Education Key Laboratory of Protein Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
- Zhejiang Provincial Key Laboratory of Applied Enzymology, Yangtze Delta Region Institute of Tsinghua University, 705 Yatai Road, Jiaxing 314006, China
- Taizhou Innovation Center, Yangtze Delta Region Institute of Tsinghua University, Jiaxing 318000, China
| |
Collapse
|
1092
|
Sharma A, Yadav A, Nandy A, Ghatak S. Insight into the Functional Dynamics and Challenges of Exosomes in Pharmaceutical Innovation and Precision Medicine. Pharmaceutics 2024; 16:709. [PMID: 38931833 PMCID: PMC11206934 DOI: 10.3390/pharmaceutics16060709] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Revised: 05/13/2024] [Accepted: 05/16/2024] [Indexed: 06/28/2024] Open
Abstract
Of all the numerous nanosized extracellular vesicles released by a cell, the endosomal-originated exosomes are increasingly recognized as potential therapeutics, owing to their inherent stability, low immunogenicity, and targeted delivery capabilities. This review critically evaluates the transformative potential of exosome-based modalities across pharmaceutical and precision medicine landscapes. Because of their precise targeted biomolecular cargo delivery, exosomes are posited as ideal candidates in drug delivery, enhancing regenerative medicine strategies, and advancing diagnostic technologies. Despite the significant market growth projections of exosome therapy, its utilization is encumbered by substantial scientific and regulatory challenges. These include the lack of universally accepted protocols for exosome isolation and the complexities associated with navigating the regulatory environment, particularly the guidelines set forth by the U.S. Food and Drug Administration (FDA). This review presents a comprehensive overview of current research trajectories aimed at addressing these impediments and discusses prospective advancements that could substantiate the clinical translation of exosomal therapies. By providing a comprehensive analysis of both the capabilities and hurdles inherent to exosome therapeutic applications, this article aims to inform and direct future research paradigms, thereby fostering the integration of exosomal systems into mainstream clinical practice.
Collapse
Affiliation(s)
| | | | | | - Subhadip Ghatak
- McGowan Institute for Regenerative Medicine, Department of Surgery, University of Pittsburgh, Pittsburgh, PA 15219, USA; (A.S.); (A.Y.); (A.N.)
| |
Collapse
|
1093
|
Di Niro L, Linders AC, Glynn T, Pegtel DM, Siderius M, Crudden C, Smit MJ. G protein-coupled receptors: a gateway to targeting oncogenic EVs? EXTRACELLULAR VESICLES AND CIRCULATING NUCLEIC ACIDS 2024; 5:233-248. [PMID: 39698539 PMCID: PMC11648488 DOI: 10.20517/evcna.2024.10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 04/29/2024] [Accepted: 05/14/2024] [Indexed: 12/20/2024]
Abstract
Dysregulated intercellular communication is a key feature driving cancer progression. Recently, extracellular vesicles (EVs) have added a new channel to this dense communication network. Despite solid evidence that EVs are central mediators of dysregulated signaling in onco-pathological settings, this has yet to be translated into clinically actionable strategies. The heterogeneity of EV cargo molecules, plasticity of biogenesis routes, and large overlap with their role in physiological communication, complicate a potential targeting strategy. However, recent work has linked EV biology to perhaps the "most druggable" proteins - G protein-coupled receptors (GPCRs). GPCR targeting accounts for ~60% of drugs in development and more than a third of all currently approved drugs, spanning almost all areas of medicine. Although several GPCRs have been linked to cancer initiation and progression, relatively few agents have made it into oncological regimes, suggesting that their potential is underexploited. Herein, we examine the molecular mechanisms linking GPCRs to EV communication in cancer settings. We propose that GPCRs hold potential in the search for EV-targeting in oncology.
Collapse
Affiliation(s)
- Lotte Di Niro
- Department of Chemistry and Pharmaceutical Sciences, Division of Medicinal Chemistry, Amsterdam Institute for Molecular and Life Sciences, Vrije Universiteit Amsterdam, Amsterdam 1081 HV, The Netherlands
| | - Amber C. Linders
- Department of Pathology, Cancer Center Amsterdam, Amsterdam UMC location Vrije Universiteit Amsterdam, Amsterdam 1081 HV, The Netherlands
| | - Thomas Glynn
- Department of Chemistry and Pharmaceutical Sciences, Division of Medicinal Chemistry, Amsterdam Institute for Molecular and Life Sciences, Vrije Universiteit Amsterdam, Amsterdam 1081 HV, The Netherlands
| | - D. Michiel Pegtel
- Department of Pathology, Cancer Center Amsterdam, Amsterdam UMC location Vrije Universiteit Amsterdam, Amsterdam 1081 HV, The Netherlands
| | - Marco Siderius
- Department of Chemistry and Pharmaceutical Sciences, Division of Medicinal Chemistry, Amsterdam Institute for Molecular and Life Sciences, Vrije Universiteit Amsterdam, Amsterdam 1081 HV, The Netherlands
| | - Caitrin Crudden
- Department of Chemistry and Pharmaceutical Sciences, Division of Medicinal Chemistry, Amsterdam Institute for Molecular and Life Sciences, Vrije Universiteit Amsterdam, Amsterdam 1081 HV, The Netherlands
- Department of Pathology, Cancer Center Amsterdam, Amsterdam UMC location Vrije Universiteit Amsterdam, Amsterdam 1081 HV, The Netherlands
| | - Martine J. Smit
- Department of Chemistry and Pharmaceutical Sciences, Division of Medicinal Chemistry, Amsterdam Institute for Molecular and Life Sciences, Vrije Universiteit Amsterdam, Amsterdam 1081 HV, The Netherlands
| |
Collapse
|
1094
|
Zhou X, Xu S, Zhang Z, Tang M, Meng Z, Peng Z, Liao Y, Yang X, Nüssler AK, Liu L, Yang W. Gouqi-derived nanovesicles (GqDNVs) inhibited dexamethasone-induced muscle atrophy associating with AMPK/SIRT1/PGC1α signaling pathway. J Nanobiotechnology 2024; 22:276. [PMID: 38778385 PMCID: PMC11112783 DOI: 10.1186/s12951-024-02563-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 05/16/2024] [Indexed: 05/25/2024] Open
Abstract
With the increasing trend of global aging, sarcopenia has become a significant public health issue. Goji berry, also known as "Gou qi zi" in China, is a traditional Chinese herb that can enhance the structure and function of muscles and bones. Otherwise, previous excellent publications illustrated that plant-derived exosome-like nanoparticles can exert good bioactive functions in different aging or disease models. Thus, we issued the hypothesis that Gouqi-derived nanovesicles (GqDNVs) may also have the ability to improve skeletal muscle health, though the effect and its mechanism need to be explored. Hence, we have extracted GqDNVs from fresh berries of Lycium barbarum L. (goji) and found that the contents of GqDNVs are rich in saccharides and lipids. Based on the pathway annotations and predictions in non-targeted metabolome analysis, GqDNVs are tightly associated with the pathways in metabolism. In muscle atrophy model mice, intramuscular injection of GqDNVs improves the cross-sectional area of the quadriceps muscle, grip strength and the AMPK/SIRT1/PGC1α pathway expression. After separately inhibiting AMPK or PGC1α in C2C12 cells with dexamethasone administration, we have found that the activated AMPK plays the chief role in improving cell proliferation induced by GqDNVs. Furthermore, the energy-targeted metabolome analysis in the quadriceps muscle demonstrates that the GqDNVs up-regulate the metabolism of amino sugar and nucleotide sugar, autophagy and oxidative phosphorylation process, which indicates the activation of muscle regeneration. Besides, the Spearman rank analysis shows close associations between the quality and function of skeletal muscle, metabolites and expression levels of AMPK and SIRT1. In this study, we provide a new founding that GqDNVs can improve the quality and function of skeletal muscle accompanying the activated AMPK/SIRT1/PGC1α signaling pathway. Therefore, GqDNVs have the effect of anti-aging skeletal muscle as a potential adjuvant or complementary method or idea in future therapy and research.
Collapse
Affiliation(s)
- Xiaolei Zhou
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan, 430030, China
- Department of Nutrition and Food Hygiene and MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan, 430030, China
| | - Shiyin Xu
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan, 430030, China
- Department of Nutrition and Food Hygiene and MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan, 430030, China
| | - Zixuan Zhang
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan, 430030, China
- Department of Nutrition and Food Hygiene and MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan, 430030, China
| | - Mingmeng Tang
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan, 430030, China
- Department of Nutrition and Food Hygiene and MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan, 430030, China
| | - Zitong Meng
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan, 430030, China
- Department of Nutrition and Food Hygiene and MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan, 430030, China
| | - Zhao Peng
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan, 430030, China
- Department of Nutrition and Food Hygiene and MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan, 430030, China
| | - Yuxiao Liao
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan, 430030, China
- Department of Nutrition and Food Hygiene and MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan, 430030, China
| | - Xuefeng Yang
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan, 430030, China
- Department of Nutrition and Food Hygiene and MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan, 430030, China
| | - Andreas K Nüssler
- Department of Traumatology, BG Trauma Center, University of Tübingen, Schnarrenbergstr. 95, 72076, Tübingen, Germany
| | - Liegang Liu
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan, 430030, China
- Department of Nutrition and Food Hygiene and MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan, 430030, China
| | - Wei Yang
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan, 430030, China.
- Department of Nutrition and Food Hygiene and MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan, 430030, China.
| |
Collapse
|
1095
|
Weber B, Ritter A, Han J, Schaible I, Sturm R, Relja B, Huber-Lang M, Hildebrand F, Pallas C, Widera M, Henrich D, Marzi I, Leppik L. Development of a Sampling and Storage Protocol of Extracellular Vesicles (EVs)-Establishment of the First EV Biobank for Polytraumatized Patients. Int J Mol Sci 2024; 25:5645. [PMID: 38891833 PMCID: PMC11172154 DOI: 10.3390/ijms25115645] [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: 03/21/2024] [Revised: 05/17/2024] [Accepted: 05/20/2024] [Indexed: 06/21/2024] Open
Abstract
In the last few years, several studies have emphasized the existence of injury-specific EV "barcodes" that could have significant importance for the precise diagnosis of different organ injuries in polytrauma patients. To expand the research potential of the NTF (network trauma research) biobank of polytraumatized patients, the NTF research group decided to further establish a biobank for EVs. However, until now, the protocols for the isolation, characterization, and storage of EVs for biobank purposes have not been conceptualized. Plasma and serum samples from healthy volunteers (n = 10) were used. Three EV isolation methods of high relevance for the work with patients' samples (ultracentrifugation, size exclusion chromatography, and immune magnetic bead-based isolation) were compared. EVs were quantified using nanoparticle tracking analysis, EV proteins, and miRNAs. The effects of different isolation solutions; the long storage of samples (up to 3 years); and the sensibility of EVs to serial freezing-thawing cycles and different storage conditions (RT, 4/-20/-80 °C, dry ice) were evaluated. The SEC isolation method was considered the most suitable for EV biobanking. We did not find any difference in the quantity of EVs between serum and plasma-EVs. The importance of particle-free PBS as an isolation solution was confirmed. Plasma that has been frozen for a long time can also be used as a source of EVs. Serial freezing-thawing cycles were found to affect the mean size of EVs but not their amount. The storage of EV samples for 5 days on dry ice significantly reduced the EV protein concentration.
Collapse
Affiliation(s)
- Birte Weber
- Department of Trauma-, Hand- and Reconstructive Surgery, University Hospital Frankfurt, Goethe University, 60486 Frankfurt am Main, Germany (L.L.)
| | - Aileen Ritter
- Department of Trauma-, Hand- and Reconstructive Surgery, University Hospital Frankfurt, Goethe University, 60486 Frankfurt am Main, Germany (L.L.)
| | - Jiaoyan Han
- Department of Trauma-, Hand- and Reconstructive Surgery, University Hospital Frankfurt, Goethe University, 60486 Frankfurt am Main, Germany (L.L.)
| | - Inna Schaible
- Department of Trauma-, Hand- and Reconstructive Surgery, University Hospital Frankfurt, Goethe University, 60486 Frankfurt am Main, Germany (L.L.)
| | - Ramona Sturm
- Department of Trauma-, Hand- and Reconstructive Surgery, University Hospital Frankfurt, Goethe University, 60486 Frankfurt am Main, Germany (L.L.)
| | - Borna Relja
- Translational and Experimental Trauma Research, Department of Trauma, Hand, Plastic and Reconstructive Surgery, University Hospital Ulm, 89081 Ulm, Germany
| | - Markus Huber-Lang
- Institute of Clinical and Experimental Trauma-Immunology, University Hospital Ulm, 89081 Ulm, Germany
| | - Frank Hildebrand
- Department of Trauma and Reconstructive Surgery, University Hospital RWTH Aachen, 52074 Aachen, Germany
| | - Christiane Pallas
- Institute for Medical Virology, University Hospital Frankfurt, Goethe University, 60596 Frankfurt am Main, Germany
| | - Marek Widera
- Institute for Medical Virology, University Hospital Frankfurt, Goethe University, 60596 Frankfurt am Main, Germany
| | - Dirk Henrich
- Department of Trauma-, Hand- and Reconstructive Surgery, University Hospital Frankfurt, Goethe University, 60486 Frankfurt am Main, Germany (L.L.)
| | - Ingo Marzi
- Department of Trauma-, Hand- and Reconstructive Surgery, University Hospital Frankfurt, Goethe University, 60486 Frankfurt am Main, Germany (L.L.)
| | - Liudmila Leppik
- Department of Trauma-, Hand- and Reconstructive Surgery, University Hospital Frankfurt, Goethe University, 60486 Frankfurt am Main, Germany (L.L.)
| |
Collapse
|
1096
|
Cetinkaya H, Kongsomros S, Nommsen-Rivers L, Morrow AL, Chutipongtanate S. Which casein micelle removal method is suitable for studies of human milk extracellular vesicles? A systematic comparison of four different treatments for casein depletion before extracellular vesicle isolation from human milk. EXTRACELLULAR VESICLES AND CIRCULATING NUCLEIC ACIDS 2024; 5:221-232. [PMID: 39698537 PMCID: PMC11648521 DOI: 10.20517/evcna.2024.02] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 04/16/2024] [Accepted: 05/14/2024] [Indexed: 12/20/2024]
Abstract
Aim: This study aimed to systematically compare four casein micelle removal methods on the particle and protein characteristics of the isolated human milk EVs. Methods: The defatted milk was treated with 1% sodium citrate, 20 mM ethylenediaminetetraacetic acid (EDTA), 1% acetic acid, or 1% chymosin/calcium chloride for 30 min at 4 °C to remove casein micelles. EV isolation was performed using qEV size exclusion chromatography. Milk turbidity at the optical density 350 nm and dot immunoblot with casein antibody were applied to monitor the qEV fractions. Particle analyses were performed using transmission electron microscopy (TEM) and nanoparticle tracking analysis (NTA). The enrichment of human milk EV markers, i.e., tetraspanins, Alix, lactadherin, butyrophilin, and xanthine dehydrogenase, and casein depletion capabilities were evaluated by proteomics and immunoblotting. Results: Compared to the untreated condition, sodium citrate and EDTA decreased milk turbidity by disrupting casein micelles, while acetic acid and chymosin removed them by inducing precipitation/coagulation. All treatments shifted casein immunoreactivity in the qEV fractions from large micelles (the exclusion volume) to small molecular sizes (gel-infiltrated fractions). Acidification affected human milk EV morphology, while EDTA, acetic acid, and chymosin methods slightly altered EV particle numbers. Different casein micelle removal methods confer different degrees of human milk EV marker enrichment and casein depletion. The method performances could be ranked as follows: chymosin > EDTA > acetic acid > sodium citrate. Conclusion: Our findings suggest that chymosin and EDTA should be considered as the method of choice for casein micelle removal in future studies involving human milk EV isolation and characterization.
Collapse
Affiliation(s)
- Hatice Cetinkaya
- MILCH and Novel Therapeutics Lab, Division of Epidemiology, Department of Environmental and Public Health Sciences, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
- Authors contributed equally
| | - Supasek Kongsomros
- MILCH and Novel Therapeutics Lab, Division of Epidemiology, Department of Environmental and Public Health Sciences, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
- Authors contributed equally
| | | | - Ardythe L. Morrow
- MILCH and Novel Therapeutics Lab, Division of Epidemiology, Department of Environmental and Public Health Sciences, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Somchai Chutipongtanate
- MILCH and Novel Therapeutics Lab, Division of Epidemiology, Department of Environmental and Public Health Sciences, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| |
Collapse
|
1097
|
Khan NLA, Muhandiram S, Dissanayake K, Godakumara K, Midekessa G, Andronowska A, Heath PR, Kodithuwakku S, Hart AR, Fazeli A. Effect of 3D and 2D cell culture systems on trophoblast extracellular vesicle physico-chemical characteristics and potency. Front Cell Dev Biol 2024; 12:1382552. [PMID: 38835509 PMCID: PMC11148233 DOI: 10.3389/fcell.2024.1382552] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 04/30/2024] [Indexed: 06/06/2024] Open
Abstract
The growing understanding of the role of extracellular vesicles (EVs) in embryo-maternal communication has sparked considerable interest in their therapeutic potential within assisted reproductive technology, particularly in enhancing implantation success. However, the major obstacle remains the large-scale production of EVs, and there is still a gap in understanding how different culture systems affect the characteristics of the EVs. In the current study, trophoblast analogue human chorionic carcinoma cell line was cultivated in both conventional monolayer culture (2D) and as spheroids in suspension culture (3D) and how the cell growth environment affects the physical, biochemical and cellular signalling properties of EVs produced by them was studied. Interestingly, the 3D system was more active in secreting EVs compared to the 2D system, while no significant differences were observed in terms of morphology, size, and classical EV protein marker expression between EVs derived from the two culture systems. There were substantial differences in the proteomic cargo profile and cellular signalling potency of EVs derived from the two culture systems. Notably, 2D EVs were more potent in inducing a cellular response in endometrial epithelial cells (EECs) compared to 3D EVs. Therefore, it is essential to recognize that the biological activity of EVs depends not only on the cell of origin but also on the cellular microenvironment of the parent cell. In conclusion, caution is warranted when selecting an EV production platform, especially for assessing the functional and therapeutic potential of EVs through in vitro studies.
Collapse
Affiliation(s)
- Norhayati Liaqat Ali Khan
- Division of Clinical Medicine, School of Medicine and Population Health, The Medical School, University of Sheffield, Sheffield, United Kingdom
- Centre of Preclinical Science Studies, Faculty of Dentistry, University Teknologi MARA (UiTM), Sg. Buloh, Selangor, Malaysia
| | - Subhashini Muhandiram
- Institute of Veterinary Medicine and Animal Sciences, Estonian University of Life Sciences, Tartu, Estonia
| | - Keerthie Dissanayake
- Institute of Veterinary Medicine and Animal Sciences, Estonian University of Life Sciences, Tartu, Estonia
- Department of Pathophysiology, Institute of Biomedicine and Translational Medicine, Faculty of Medicine, University of Tartu, Tartu, Estonia
| | - Kasun Godakumara
- Institute of Veterinary Medicine and Animal Sciences, Estonian University of Life Sciences, Tartu, Estonia
- Department of Pathophysiology, Institute of Biomedicine and Translational Medicine, Faculty of Medicine, University of Tartu, Tartu, Estonia
| | - Getnet Midekessa
- Institute of Veterinary Medicine and Animal Sciences, Estonian University of Life Sciences, Tartu, Estonia
- Department of Pathophysiology, Institute of Biomedicine and Translational Medicine, Faculty of Medicine, University of Tartu, Tartu, Estonia
| | - Aneta Andronowska
- Department of Hormonal Action Mechanisms, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn, Poland
| | - Paul R Heath
- Division of Clinical Medicine, School of Medicine and Population Health, The Medical School, University of Sheffield, Sheffield, United Kingdom
| | - Suranga Kodithuwakku
- Institute of Veterinary Medicine and Animal Sciences, Estonian University of Life Sciences, Tartu, Estonia
- Department of Animal Science, Faculty of Agriculture, University of Peradeniya, Peradeniya, Sri Lanka
| | - Amber Rose Hart
- Division of Clinical Medicine, School of Medicine and Population Health, The Medical School, University of Sheffield, Sheffield, United Kingdom
| | - Alireza Fazeli
- Division of Clinical Medicine, School of Medicine and Population Health, The Medical School, University of Sheffield, Sheffield, United Kingdom
- Institute of Veterinary Medicine and Animal Sciences, Estonian University of Life Sciences, Tartu, Estonia
- Department of Pathophysiology, Institute of Biomedicine and Translational Medicine, Faculty of Medicine, University of Tartu, Tartu, Estonia
| |
Collapse
|
1098
|
Menjivar NG, Oropallo J, Gebremedhn S, Souza LA, Gad A, Puttlitz CM, Tesfaye D. MicroRNA Nano-Shuttles: Engineering Extracellular Vesicles as a Cutting-Edge Biotechnology Platform for Clinical Use in Therapeutics. Biol Proced Online 2024; 26:14. [PMID: 38773366 PMCID: PMC11106895 DOI: 10.1186/s12575-024-00241-6] [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: 04/04/2024] [Accepted: 04/30/2024] [Indexed: 05/23/2024] Open
Abstract
Extracellular vesicles (EVs) are nano-sized, membranous transporters of various active biomolecules with inflicting phenotypic capabilities, that are naturally secreted by almost all cells with a promising vantage point as a potential leading drug delivery platform. The intrinsic characteristics of their low toxicity, superior structural stability, and cargo loading capacity continue to fuel a multitude of research avenues dedicated to loading EVs with therapeutic and diagnostic cargos (pharmaceutical compounds, nucleic acids, proteins, and nanomaterials) in attempts to generate superior natural nanoscale delivery systems for clinical application in therapeutics. In addition to their well-known role in intercellular communication, EVs harbor microRNAs (miRNAs), which can alter the translational potential of receiving cells and thus act as important mediators in numerous biological and pathological processes. To leverage this potential, EVs can be structurally engineered to shuttle therapeutic miRNAs to diseased recipient cells as a potential targeted 'treatment' or 'therapy'. Herein, this review focuses on the therapeutic potential of EV-coupled miRNAs; summarizing the biogenesis, contents, and function of EVs, as well as providing both a comprehensive discussion of current EV loading techniques and an update on miRNA-engineered EVs as a next-generation platform piloting benchtop studies to propel potential clinical translation on the forefront of nanomedicine.
Collapse
Affiliation(s)
- Nico G Menjivar
- Animal Reproduction and Biotechnology Laboratory (ARBL), Department of Biomedical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, 80523, USA
| | - Jaiden Oropallo
- Orthopaedic Bioengineering Research Laboratory (OBRL), Translational Medicine Institute (TMI), Department of Mechanical Engineering, Colorado State University, Fort Collins, CO, 80523, USA
- Orthopaedic Research Center (ORC), Translational Medicine Institute (TMI), Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Science, Colorado State University, Fort Collins, CO, 80523, USA
| | - Samuel Gebremedhn
- Animal Reproduction and Biotechnology Laboratory (ARBL), Department of Biomedical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, 80523, USA
- J.R. Simplot Company, 1099 W. Front St, Boise, ID, 83702, USA
| | - Luca A Souza
- Animal Reproduction and Biotechnology Laboratory (ARBL), Department of Biomedical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, 80523, USA
- Department of Veterinary Medicine, College of Animal Science and Food Engineering, University of São Paulo, 225 Av. Duque de Caxias Norte, Pirassununga, SP, 13635-900, Brazil
| | - Ahmed Gad
- Animal Reproduction and Biotechnology Laboratory (ARBL), Department of Biomedical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, 80523, USA
- Department of Animal Production, Faculty of Agriculture, Cairo University, Giza, 12613, Egypt
| | - Christian M Puttlitz
- Orthopaedic Bioengineering Research Laboratory (OBRL), Translational Medicine Institute (TMI), Department of Mechanical Engineering, Colorado State University, Fort Collins, CO, 80523, USA
| | - Dawit Tesfaye
- Animal Reproduction and Biotechnology Laboratory (ARBL), Department of Biomedical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, 80523, USA.
| |
Collapse
|
1099
|
Hindle J, Williams A, Kim Y, Kim D, Patil K, Khatkar P, Osgood Q, Nelson C, Routenberg DA, Howard M, Liotta LA, Kashanchi F, Branscome H. hTERT-Immortalized Mesenchymal Stem Cell-Derived Extracellular Vesicles: Large-Scale Manufacturing, Cargo Profiling, and Functional Effects in Retinal Epithelial Cells. Cells 2024; 13:861. [PMID: 38786083 PMCID: PMC11120263 DOI: 10.3390/cells13100861] [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: 04/13/2024] [Revised: 05/07/2024] [Accepted: 05/14/2024] [Indexed: 05/25/2024] Open
Abstract
As the economic burden associated with vision loss and ocular damage continues to rise, there is a need to explore novel treatment strategies. Extracellular vesicles (EVs) are enriched with various biological cargo, and there is abundant literature supporting the reparative and immunomodulatory properties of stem cell EVs across a broad range of pathologies. However, one area that requires further attention is the reparative effects of stem cell EVs in the context of ocular damage. Additionally, most of the literature focuses on EVs isolated from primary stem cells; the use of EVs isolated from human telomerase reverse transcriptase (hTERT)-immortalized stem cells has not been thoroughly examined. Using our large-scale EV-manufacturing platform, we reproducibly manufactured EVs from hTERT-immortalized mesenchymal stem cells (MSCs) and employed various methods to characterize and profile their associated cargo. We also utilized well-established cell-based assays to compare the effects of these EVs on both healthy and damaged retinal pigment epithelial cells. To the best of our knowledge, this is the first study to establish proof of concept for reproducible, large-scale manufacturing of hTERT-immortalized MSC EVs and to investigate their potential reparative properties against damaged retinal cells. The results from our studies confirm that hTERT-immortalized MSC EVs exert reparative effects in vitro that are similar to those observed in primary MSC EVs. Therefore, hTERT-immortalized MSCs may represent a more consistent and reproducible platform than primary MSCs for generating EVs with therapeutic potential.
Collapse
Affiliation(s)
| | - Anastasia Williams
- Laboratory of Molecular Virology, School of Systems Biology, George Mason University, Manassas, VA 20110, USA (K.P.)
| | - Yuriy Kim
- Laboratory of Molecular Virology, School of Systems Biology, George Mason University, Manassas, VA 20110, USA (K.P.)
| | | | - Kajal Patil
- Laboratory of Molecular Virology, School of Systems Biology, George Mason University, Manassas, VA 20110, USA (K.P.)
| | - Pooja Khatkar
- Laboratory of Molecular Virology, School of Systems Biology, George Mason University, Manassas, VA 20110, USA (K.P.)
| | | | - Collin Nelson
- Meso Scale Diagnostics, L.L.C., Rockville, MD 20850, USA (D.A.R.)
| | | | - Marissa Howard
- Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, VA 20110, USA
| | - Lance A. Liotta
- Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, VA 20110, USA
| | - Fatah Kashanchi
- Laboratory of Molecular Virology, School of Systems Biology, George Mason University, Manassas, VA 20110, USA (K.P.)
| | - Heather Branscome
- ATCC, Manassas, VA 20110, USA
- Laboratory of Molecular Virology, School of Systems Biology, George Mason University, Manassas, VA 20110, USA (K.P.)
| |
Collapse
|
1100
|
O'Toole HJ, Lowe N, Arun V, Kolesov AV, Palmieri TL, Tran NK, Carney RP. Plasma-derived Extracellular Vesicles (EVs) as Biomarkers of Sepsis in Burn Patients via Label-free Raman Spectroscopy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.14.593634. [PMID: 38798662 PMCID: PMC11118394 DOI: 10.1101/2024.05.14.593634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Sepsis following burn trauma is a global complication with high mortality, with ~60% of burn patient deaths resulting from infectious complications. Sepsis diagnosis is complicated by confounding clinical manifestations of the burn injury, and current biomarkers markers lack the sensitivity and specificity required for prompt treatment. Circulating extracellular vesicles (EVs) from patient liquid biopsy as biomarkers of sepsis due to their release by pathogens from bacterial biofilms and roles in subsequent immune response. This study applies Raman spectroscopy to patient plasma derived EVs for rapid, sensitive, and specific detection of sepsis in burn patients, achieving 97.5% sensitivity and 90.0% specificity. Furthermore, spectral differences between septic and non-septic burn patient EVs could be traced to specific glycoconjugates of bacterial strains associated with sepsis morbidity. This work illustrates the potential application of EVs as biomarkers in clinical burn trauma care, and establishes Raman analysis as a fast, label-free method to specifically identify features of bacterial EVs relevant to infection amongst the host background.
Collapse
Affiliation(s)
- Hannah J O'Toole
- Department of Biomedical Engineering, University of California, Davis, 1 Shields Ave, Davis., CA 95616, USA
| | - Neona Lowe
- Department of Biomedical Engineering, University of California, Davis, 1 Shields Ave., Davis, CA 95616, USA
| | - Vishalakshi Arun
- Department of Neurobiology, Physiology, and Behavior, University of California, Davis, 1 Shields Ave., Davis, CA 95616, USA
| | - Anna V Kolesov
- Department of Biomedical Engineering, University of California, Davis, 1 Shields Ave., Davis, CA 95616, USA
| | - Tina L Palmieri
- Division of Burn Surgery & Reconstruction, Department of Surgery, University of California, Davis Health, Firefighters Burn Institute Regional Burn Center, 2315 X Street, Sacramento, CA 95616, USA; Shriners Hospitals for Children Northern California, 2425 Stockton Blvd., Sacramento, CA 95817, USA
| | - Nam K Tran
- Department of Pathology and Laboratory Medicine, University of California, Davis, 4400 V. St., Sacramento, CA 95817, USA
| | - Randy P Carney
- Department of Biomedical Engineering, University of California, Davis, 1 Shields Ave, Davis, CA 95616, USA
| |
Collapse
|