1
|
Sato S, Teramura Y, Ogawa Y, Shimizu E, Otake M, Hori K, Kamata T, Shu Y, Seta Y, Kuramochi A, Asai K, Shimizu S, Negishi K, Hirayama M. Conditioned media of stem cells from human exfoliated deciduous teeth contain factors related to extracellular matrix organization and promotes corneal epithelial wound healing. Regen Ther 2025; 29:148-161. [PMID: 40170802 PMCID: PMC11960544 DOI: 10.1016/j.reth.2025.03.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Revised: 02/27/2025] [Accepted: 03/09/2025] [Indexed: 04/03/2025] Open
Abstract
This study aimed to investigate the therapeutic potential of cell-free conditioned media (CM) from human mesenchymal stem cells (hMSCs), specifically stem cells from human exfoliated deciduous teeth (SHED), for treating ocular surface diseases. The proteomes of various hMSC-CMs were compared using cytokine array and liquid chromatography-mass spectrometry (LC-MS). Bioinformatic analysis identified key biological pathways associated with SHED-CM, immortalized SHED-CM (IM-SHED-CM), and a fractionated component of IM-SHED-CM in which low weight molecules (less than 3.5kD) were depleted. Corneal epithelial wound healing models were constructed by epithelial scraping and treated with eye drops derived from SHED-CM. For the migration assay, the human corneal epithelial cells were wounded and then incubated with SHED-CM. SHED-CM, IM-SHED-CM, and >3.5 kD fractionated component eyedrops were administered to a chronic graft-versus-host disease (cGVHD) mouse model with sever corneal epithelial damages. SHED-CM, IM-SHED-CM, and >3.5 kD fractionated component of IM-SHED-CM were enriched in factors involved in epithelial wound healing, particularly extracellular matrix (ECM) organization. Both in vitro and in vivo assays demonstrated that SHED-CM significantly enhanced corneal epithelial wound healing. Furthermore, SHED-CM-derived eye drops reduced corneal epithelial damage, inflammatory cell infiltration, and oxidative stress in the corneal epithelium and maintained the expression of limbal stem cell markers in the cGVHD mouse model. These findings suggest that SHED-CM eye drops could be a novel treatment for corneal epithelial damage, highlighting the role of bioactive factors in promoting wound healing and offering an alternative to cell-based MSC therapies for corneal wound healing.
Collapse
Affiliation(s)
- Shinri Sato
- Department of Ophthalmology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Yuji Teramura
- Cellular and Molecular Biotechnology Research Institute (CMB), National Institute of Advanced Industrial Science and Technology (AIST), AIST Tsukuba Central 5, 1-1-1 Higashi, Tsukuba, Ibaraki 305-8565, Japan
- Department of Immunology, Genetics and Pathology (IGP), Uppsala University, Dag Hammarskjölds väg 20, SE-751 85, Uppsala, Sweden
- Master's/Doctoral Program in Life Science Innovation (T-LSI), University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8577, Japan
| | - Yoko Ogawa
- Department of Ophthalmology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Eisuke Shimizu
- Department of Ophthalmology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Masato Otake
- U-Factor Co., Ltd., 1F ESCALIER Rokubancho, 7-11 Rokubancho, Chiyoda-ku, Tokyo 102-0085, Japan
| | - Keigo Hori
- U-Factor Co., Ltd., 1F ESCALIER Rokubancho, 7-11 Rokubancho, Chiyoda-ku, Tokyo 102-0085, Japan
| | - Takamitsu Kamata
- U-Factor Co., Ltd., 1F ESCALIER Rokubancho, 7-11 Rokubancho, Chiyoda-ku, Tokyo 102-0085, Japan
| | - Yujing Shu
- U-Factor Co., Ltd., 1F ESCALIER Rokubancho, 7-11 Rokubancho, Chiyoda-ku, Tokyo 102-0085, Japan
| | - Yasuhiro Seta
- Hitonowa Medical, K. PLAZA 2F, 1-7 Rokubancho, Chiyoda-ku, Tokyo 102-0085, Japan
| | - Akiko Kuramochi
- Cellular and Molecular Biotechnology Research Institute (CMB), National Institute of Advanced Industrial Science and Technology (AIST), AIST Tsukuba Central 5, 1-1-1 Higashi, Tsukuba, Ibaraki 305-8565, Japan
| | - Kazuki Asai
- Department of Ophthalmology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Shota Shimizu
- Department of Ophthalmology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Kazuno Negishi
- Department of Ophthalmology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Masatoshi Hirayama
- Department of Ophthalmology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| |
Collapse
|
2
|
Katsuba KE, Zabegina LM, Plevako DS, Gurtovenko AA, Malek AV. Targeting HER2 with DNA Aptamers for Efficient Anticancer Drug Delivery: A Combined Experimental and Computational Study. Bioconjug Chem 2025. [PMID: 40403699 DOI: 10.1021/acs.bioconjchem.5c00022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/24/2025]
Abstract
Targeted delivery of cytostatic drugs is a powerful approach to achieving tumor tissue selectivity, reducing systemic toxicity, and ultimately improving the efficacy of anticancer chemotherapy. Targeting can be achieved using a wide range of molecular ligands, with DNA aptamers being a promising representative. In this work, we employed flow cytometry, a AuNP-aptasensor, and atomic-scale computer modeling to assess the affinity of several DNA aptamers (Anti-HER2, HB5, Apt-6, HeA2_1, and HeA2_3) for human epidermal growth factor receptor 2 (HER2), which is known to be one of the factors that promote the growth of breast cancer cells. Flow cytometry showed that short aptamers (HeA2_1 and HeA2_3) had a higher affinity for HER2 on MDAMB453 cancer cells than longer aptamers (HB5, Apt-6). HER2-negative MDA-MB-231 cells served as the negative control. The HeA2_3 aptamer has a high average affinity (HeA2_3:23.6, HeA2_1:13.1, Apt-6:3.6; HB5:3.5; Anti-HER2:3.2) and a nearly Gaussian distribution across the cells, while HeA2_1 forms a fraction of cells with a relatively high fluorescence signal intensity (HeA2_1:11.6; HeA2_3:5.9; Apt-6:3.4; HB5:3.1; Anti-HER2:2.1). Most of the findings for cancer cells also hold for the HER2-positive small extracellular vesicles studied using the AuNP-aptasensor. Computer simulations confirmed that short aptamers are characterized by stronger binding to the extracellular domain of HER2. A detailed analysis of the free energy allowed us to show for the first time that tight binding to HER2 correlates with well-separated hot and cold spots on the protein surface. For the aptamers that meet these criteria (HeA2_1, HeA2_3, and Anti-HER2), favorable interactions with HER2 are driven by the local attraction of nucleotides to arginine and lysine residues of HER2 and possibly stabilized by intermolecular hydrogen bonds. For longer aptamers (Apt-6 and HB5), hot and cold spots on the HER2 surface overlap and the aptamers show much weaker binding. Overall, our findings show that binding of DNA aptamers to HER2 cannot be characterized merely by the dissociation equilibrium constant. A more sophisticated approach that combines experimental and computational methods allowed us to unlock the molecular mechanisms behind the aptamer-HER2 bindings. The results of our study also suggest that computer modeling has become a reliable and accurate tool for aptamer prescreening prior to laboratory experiments.
Collapse
Affiliation(s)
- Konstantin E Katsuba
- N.N. Petrov National Medical Research Center of Oncology, Pesochny, Leningradskaya 68, St. Petersburg 197758, Russia
| | - Lidia M Zabegina
- N.N. Petrov National Medical Research Center of Oncology, Pesochny, Leningradskaya 68, St. Petersburg 197758, Russia
| | - Daniil S Plevako
- N.N. Petrov National Medical Research Center of Oncology, Pesochny, Leningradskaya 68, St. Petersburg 197758, Russia
| | - Andrey A Gurtovenko
- Branch of Petersburg Nuclear Physics Institute named by B.P. Konstantinov of National Research Centre "Kurchatov Institute", Institute of Macromolecular Compounds, Bolshoi Prospect V.O. 31, St. Petersburg 199004, Russia
| | - Anastasia V Malek
- N.N. Petrov National Medical Research Center of Oncology, Pesochny, Leningradskaya 68, St. Petersburg 197758, Russia
| |
Collapse
|
3
|
WANG H, XIE P, QIAO X, ZHANG L. [Typical strategy and research progress of efficient isolation methods of exosomes based on affinity interaction]. Se Pu 2025; 43:413-423. [PMID: 40331606 PMCID: PMC12059992 DOI: 10.3724/sp.j.1123.2024.11004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Indexed: 05/08/2025] Open
Abstract
Exosomes form a subclass of extracellular vesicle that are secreted by most cells and found in nearly all body fluids, including blood, urine, saliva, amniotic fluid, and milk, as well as in various tissues and intercellular spaces. Exosomes have recently been recognized as crucial intercellular communication mediators, and an increasing number of studies have shown that exosomes are important liquid-biopsy tools that play irreplaceable roles in the diagnosis, prognosis, and treatment of diseases. The ability to isolate high-quality exosomes is a prerequisite for diagnosing and subsequently treating diseases in an accurate and repeatable manner. However, efficiently isolating exosomes from complex biological samples is challenging owing to their relatively low abundances and interference from non-vesicular macromolecules (such as cell debris and proteins). To date, various isolation techniques based on the physical, chemical, and biological characteristics of exosomes have been developed. Indeed, efficient affinity-interaction-based methods have recently overcome the limitations and drawbacks of traditional exosome isolation methods and are widely used in scientific research and clinical applications. This review focuses on exosome isolation and enrichment, and systematically reviews recent research progress on efficient isolation methods based on affinity interactions. Developmental prospects of exosome isolation and enrichment directions are analyzed with the aim of providing a reference for the construction and use of new exosome-isolation strategies.
Collapse
|
4
|
LaRocca TJ, Lark DS. Mapping Organism-wide Single Cell mRNA Expression Linked to Extracellular Vesicle Biogenesis, Secretion, and Cargo. FUNCTION 2025; 6:zqaf005. [PMID: 39863422 PMCID: PMC11931722 DOI: 10.1093/function/zqaf005] [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: 10/25/2024] [Revised: 01/06/2025] [Accepted: 01/23/2025] [Indexed: 01/27/2025] Open
Abstract
Extracellular vesicles (EVs) are functional lipid-bound nanoparticles trafficked between cells and found in every biofluid. It is widely claimed that EVs can be secreted by every cell, but the quantity and composition of these EVs can differ greatly among cell types and tissues. Defining this heterogeneity has broad implications for EV-based communication in health and disease. Recent discoveries have linked single-cell EV secretion to the expression of genes encoding EV machinery and cargo. To gain insight at single-cell resolution across an entire organism, we compared the abundance, variance, and co-expression of 67 genes involved in EV biogenesis and secretion, or carried as cargo, across >44 000 cells obtained from 117 cell populations in the Tabula Muris. Our analysis provides both novel holistic and cell population-specific insight into EV biology. The highest overall expression of EV genes occurs in secretory cells of the pancreas and perhaps more surprisingly, multiple non-neuronal cell populations of the brain. We find that the most abundant EV genes encode the most abundant EV cargo proteins (tetraspanins and syndecans), but these genes are highly differentially expressed across functionally distinct cell populations. Expression variance identifies dynamic and constitutively expressed EV genes while co-expression analysis reveals novel insights into cell population-specific coordination of expression. Results of our analysis illustrate the diverse transcriptional regulation of EV genes which could be useful for predicting how individual cell populations might communicate via EVs to influence health and disease.
Collapse
Affiliation(s)
- Thomas J LaRocca
- Department of Health and Exercise Science, College of Health and Human Sciences, Colorado State University, Fort Collins, CO 80521, USA
- Columbine Health Systems Center for Healthy Aging, Colorado State University, Fort Collins, CO 80521, USA
| | - Daniel S Lark
- Department of Health and Exercise Science, College of Health and Human Sciences, Colorado State University, Fort Collins, CO 80521, USA
- Columbine Health Systems Center for Healthy Aging, Colorado State University, Fort Collins, CO 80521, USA
| |
Collapse
|
5
|
Puagsopa J, Tongviseskul N, Jaroentomeechai T, Meksiriporn B. Recent Progress in Developing Extracellular Vesicles as Nanovehicles to Deliver Carbohydrate-Based Therapeutics and Vaccines. Vaccines (Basel) 2025; 13:285. [PMID: 40266147 PMCID: PMC11946770 DOI: 10.3390/vaccines13030285] [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: 01/31/2025] [Revised: 02/22/2025] [Accepted: 03/04/2025] [Indexed: 04/24/2025] Open
Abstract
Cell-derived, nanoscale extracellular vesicles (EVs) have emerged as promising tools in diagnostic, therapeutic, and vaccine applications. Their unique properties including the capability to encapsulate diverse molecular cargo as well as the versatility in surface functionalization make them ideal candidates for safe and effective vehicles to deliver a range of biomolecules including gene editing cassettes, therapeutic proteins, glycans, and glycoconjugate vaccines. In this review, we discuss recent advances in the development of EVs derived from mammalian and bacterial cells for use in a delivery of carbohydrate-based protein therapeutics and vaccines. We highlight key innovations in EVs' molecular design, characterization, and deployment for treating diseases including Alzheimer's disease, infectious diseases, and cancers. We discuss challenges for their clinical translation and provide perspectives for future development of EVs within biopharmaceutical research and the clinical translation landscape.
Collapse
Affiliation(s)
- Japigorn Puagsopa
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, FL 32610, USA;
| | - Niksa Tongviseskul
- Department of Biology, School of Science, King Mongkut’s Institute of Technology Ladkrabang, Bangkok 10520, Thailand;
| | - Thapakorn Jaroentomeechai
- Copenhagen Center for Glycomics, Departments of Cellular and Molecular Medicine, Faculty of Health Sciences, University of Copenhagen, Blegdamsvej 3, 2200 Copenhagen, Denmark
| | - Bunyarit Meksiriporn
- Department of Biology, School of Science, King Mongkut’s Institute of Technology Ladkrabang, Bangkok 10520, Thailand;
| |
Collapse
|
6
|
Holcar M, Marić I, Tertel T, Goričar K, Čegovnik Primožič U, Černe D, Giebel B, Lenassi M. Comprehensive Phenotyping of Extracellular Vesicles in Plasma of Healthy Humans - Insights Into Cellular Origin and Biological Variation. J Extracell Vesicles 2025; 14:e70039. [PMID: 39834131 PMCID: PMC11746918 DOI: 10.1002/jev2.70039] [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: 07/04/2024] [Revised: 12/21/2024] [Accepted: 01/02/2025] [Indexed: 01/22/2025] Open
Abstract
Despite immense interest in biomarker applications of extracellular vesicles (EVs) from blood, our understanding of circulating EVs under physiological conditions in healthy humans remains limited. Using imaging and multiplex bead-based flow cytometry, we comprehensively quantified circulating EVs with respect to their cellular origin in a large cohort of healthy blood donors. We assessed coefficients of variations to characterize their biological variation and explored demographic, clinical, and lifestyle factors contributing to observed variation. Cell-specific circulating EV subsets show a wide range of concentrations that do not correlate with cell-of-origin concentrations in blood, suggesting steady-state EV subset concentrations are regulated by complex mechanisms, which differ even for EV subsets from the same cell type. Interestingly, tetraspanin+ circulating EVs largely originate from platelets and to a lesser extent from lymphocytes. Principal component analysis (PCA) and association analyses demonstrate high biological inter-individual variation in circulating EVs across healthy humans, which are only partly explained by the influence of sex, menopausal status, age and smoking on specific circulating EV and/or tetraspanin+ circulating EV subsets. No global influence of the explored subject's factors on circulating EVs was detected. Our findings provide the first comprehensive, quantitative data towards the cell-origin atlas of plasma EVs, with important implications in the clinical use of EVs as biomarkers.
Collapse
Affiliation(s)
- Marija Holcar
- Institute of Biochemistry and Molecular Genetics, Faculty of MedicineUniversity of LjubljanaLjubljanaSlovenia
| | - Ivica Marić
- Department of ImmunohaematologyBlood Transfusion Centre of SloveniaLjubljanaSlovenia
- Faculty of MedicineUniversity of LjubljanaLjubljanaSlovenia
| | - Tobias Tertel
- Institute for Transfusion MedicineUniversity Hospital Essen, University of Duisburg‐EssenEssenGermany
| | - Katja Goričar
- Institute of Biochemistry and Molecular Genetics, Faculty of MedicineUniversity of LjubljanaLjubljanaSlovenia
| | - Urška Čegovnik Primožič
- Clinical Institute for Clinical Chemistry and BiochemistryUniversity Medical Centre LjubljanaLjubljanaSlovenia
| | - Darko Černe
- Clinical Institute for Clinical Chemistry and BiochemistryUniversity Medical Centre LjubljanaLjubljanaSlovenia
| | - Bernd Giebel
- Institute for Transfusion MedicineUniversity Hospital Essen, University of Duisburg‐EssenEssenGermany
| | - Metka Lenassi
- Institute of Biochemistry and Molecular Genetics, Faculty of MedicineUniversity of LjubljanaLjubljanaSlovenia
| |
Collapse
|
7
|
Geyer PE, Hornburg D, Pernemalm M, Hauck SM, Palaniappan KK, Albrecht V, Dagley LF, Moritz RL, Yu X, Edfors F, Vandenbrouck Y, Mueller-Reif JB, Sun Z, Brun V, Ahadi S, Omenn GS, Deutsch EW, Schwenk JM. The Circulating Proteome─Technological Developments, Current Challenges, and Future Trends. J Proteome Res 2024; 23:5279-5295. [PMID: 39479990 PMCID: PMC11629384 DOI: 10.1021/acs.jproteome.4c00586] [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: 07/09/2024] [Revised: 09/26/2024] [Accepted: 09/27/2024] [Indexed: 11/02/2024]
Abstract
Recent improvements in proteomics technologies have fundamentally altered our capacities to characterize human biology. There is an ever-growing interest in using these novel methods for studying the circulating proteome, as blood offers an accessible window into human health. However, every methodological innovation and analytical progress calls for reassessing our existing approaches and routines to ensure that the new data will add value to the greater biomedical research community and avoid previous errors. As representatives of HUPO's Human Plasma Proteome Project (HPPP), we present our 2024 survey of the current progress in our community, including the latest build of the Human Plasma Proteome PeptideAtlas that now comprises 4608 proteins detected in 113 data sets. We then discuss the updates of established proteomics methods, emerging technologies, and investigations of proteoforms, protein networks, extracellualr vesicles, circulating antibodies and microsamples. Finally, we provide a prospective view of using the current and emerging proteomics tools in studies of circulating proteins.
Collapse
Affiliation(s)
- Philipp E. Geyer
- Department
of Proteomics and Signal Transduction, Max
Planck Institute of Biochemistry, 82152 Martinsried, Germany
| | - Daniel Hornburg
- Seer,
Inc., Redwood City, California 94065, United States
- Bruker
Scientific, San Jose, California 95134, United States
| | - Maria Pernemalm
- Department
of Oncology and Pathology/Science for Life Laboratory, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Stefanie M. Hauck
- Metabolomics
and Proteomics Core, Helmholtz Zentrum München
GmbH, German Research Center for Environmental Health, 85764 Oberschleissheim,
Munich, Germany
| | | | - Vincent Albrecht
- Department
of Proteomics and Signal Transduction, Max
Planck Institute of Biochemistry, 82152 Martinsried, Germany
| | - Laura F. Dagley
- The
Walter and Eliza Hall Institute for Medical Research, Parkville, VIC 3052, Australia
- Department
of Medical Biology, University of Melbourne, Parkville, VIC 3052, Australia
| | - Robert L. Moritz
- Institute
for Systems Biology, Seattle, Washington 98109, United States
| | - Xiaobo Yu
- State
Key Laboratory of Medical Proteomics, Beijing
Proteome Research Center, National Center for Protein Sciences-Beijing
(PHOENIX Center), Beijing Institute of Lifeomics, Beijing 102206, China
| | - Fredrik Edfors
- Science
for Life Laboratory, Department of Protein Science, KTH Royal Institute of Technology, 17121 Solna, Sweden
| | | | - Johannes B. Mueller-Reif
- Department
of Proteomics and Signal Transduction, Max
Planck Institute of Biochemistry, 82152 Martinsried, Germany
| | - Zhi Sun
- Institute
for Systems Biology, Seattle, Washington 98109, United States
| | - Virginie Brun
- Université Grenoble
Alpes, CEA, Leti, Clinatec, Inserm UA13
BGE, CNRS FR2048, Grenoble, France
| | - Sara Ahadi
- Alkahest, Inc., Suite
D San Carlos, California 94070, United States
| | - Gilbert S. Omenn
- Institute
for Systems Biology, Seattle, Washington 98109, United States
- Departments
of Computational Medicine & Bioinformatics, Internal Medicine,
Human Genetics and Environmental Health, University of Michigan, Ann Arbor, Michigan 48109-2218, United States
| | - Eric W. Deutsch
- Institute
for Systems Biology, Seattle, Washington 98109, United States
| | - Jochen M. Schwenk
- Science
for Life Laboratory, Department of Protein Science, KTH Royal Institute of Technology, 17121 Solna, Sweden
| |
Collapse
|
8
|
Chen HT, Yi Y, Huang WY, Wu MY, Xiong Q, Wang XR, Liu M, Wu X, Jiang GL, Zhuang HW, Chen KT, Xiong GX, Fang SB. Characterization of the components in plasma EVs unveiling the link between EVs-derived complement C3 with the severity and initial treatment response of profound sudden sensorineural hearing loss. Int Immunopharmacol 2024; 141:112944. [PMID: 39153308 DOI: 10.1016/j.intimp.2024.112944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 08/03/2024] [Accepted: 08/12/2024] [Indexed: 08/19/2024]
Abstract
BACKGROUND Sudden sensorineural hearing loss (SSNHL) is characterized by rapid, unexplained loss of hearing within a 72-hour period and exhibits a high incidence globally. Despite this, the outcomes of therapeutic interventions remain largely unpredictable, especially for those with profound hearing loss. Extracellular vesicles (EVs), nano-sized entities containing biological materials, are implicated in the development of numerous diseases. The specific relationship between EVs and both the severity and treatment effectiveness of SSNHL, however, is not well understood. METHODS This study involved the analysis of medical records from the Department of Otolaryngology (September 1, 2020 - December 31, 2022) of patients diagnosed with SSNHL according to the 2015 Guidelines for Diagnosis and Treatment of Sudden Deafness in China. Peripheral blood samples from patients with various types of SSNHL before and after treatment were collected, alongside samples from healthy volunteers serving as controls. Plasma EVs were isolated using gel rejection chromatography and analyzed for concentration, marker presence, and morphology using Nanosight, Western blot, and transmission electron microscopy (TEM), respectively. Proteomics and miRNA assessments were conducted to identify differentially expressed proteins and miRNAs in the plasma EVs of SSNHL patients and healthy volunteers. Key proteins were further validated through Western blot analysis. Enzyme-linked immunosorbent assay (ELISA) was utilized to determine the levels of complement C3 in plasma EVs, and correlation analyses were performed with audiological data pre- and post-treatment. RESULTS Plasma from SSNHL patients of varying types was collected and their EVs were successfully isolated and characterized. Proteomic analysis revealed that complement C3 levels in the plasma EVs of patients with profound SSNHL were significantly higher compared to healthy controls. Differential expression of miRNAs in plasma EVs and their related functions were also identified. The study found that the level of complement C3 in plasma EVs, but not the total plasma complement C3, positively correlated with the severity of SSNHL in patients exhibiting positive therapeutic responses, particularly in those with initially lower levels of EV-associated complement C3. After treatment, complement C3 level was decreased in patients with initially higher levels of EV-associated complement C3. No significant correlation was observed between changes in plasma EV-derived complement C3 levels and the degree of hearing loss in either responders or non-responders among patients with profound SSNHL. CONCLUSION Differential profiles of proteins and miRNAs were identified in patients with profound SSNHL. Notably, plasma EV-derived complement C3 was linked to both the severity and early treatment effectiveness of patients with profound SSNHL.
Collapse
Affiliation(s)
- Hui-Ting Chen
- Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan Road II, Guangzhou, China; Department of Otolaryngology, Head and Neck Surgery, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, China
| | - Ying Yi
- Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan Road II, Guangzhou, China
| | - Wan-Yi Huang
- Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan Road II, Guangzhou, China
| | - Min-Yu Wu
- Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan Road II, Guangzhou, China
| | - Qin Xiong
- Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan Road II, Guangzhou, China
| | - Xian-Ren Wang
- Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan Road II, Guangzhou, China
| | - Min Liu
- Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan Road II, Guangzhou, China
| | - Xuan Wu
- Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan Road II, Guangzhou, China
| | - Guang-Li Jiang
- Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan Road II, Guangzhou, China
| | - Hui-Wen Zhuang
- Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan Road II, Guangzhou, China
| | - Kai-Tian Chen
- Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan Road II, Guangzhou, China.
| | - Guan-Xia Xiong
- Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan Road II, Guangzhou, China.
| | - Shu-Bin Fang
- Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan Road II, Guangzhou, China.
| |
Collapse
|
9
|
Esparza D, Lima C, Abuelreich S, Ghaeli I, Hwang J, Oh E, Lenz A, Gu A, Jiang N, Kandeel F, Thurmond DC, Jovanovic-Talisman T. Pancreatic β-cells package double C2-like domain beta protein into extracellular vesicles via tandem C2 domains. Front Endocrinol (Lausanne) 2024; 15:1451279. [PMID: 39497805 PMCID: PMC11532064 DOI: 10.3389/fendo.2024.1451279] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 09/10/2024] [Indexed: 11/07/2024] Open
Abstract
Introduction Double C2-like domain beta (DOC2B) is a vesicle priming protein critical for glucose-stimulated insulin secretion in β-cells. Individuals with type 1 diabetes (T1D) have lower levels of DOC2B in their residual functional β-cell mass and platelets, a phenotype also observed in a mouse model of T1D. Thus, DOC2B levels could provide important information on β-cell dys(function). Objective Our objective was to evaluate the DOC2B secretome of β-cells. In addition to soluble extracellular protein, we assessed DOC2B localized within membrane-delimited nanoparticles - extracellular vesicles (EVs). Moreover, in rat clonal β-cells, we probed domains required for DOC2B sorting into EVs. Method Using Single Extracellular VEsicle Nanoscopy, we quantified EVs derived from clonal β-cells (human EndoC-βH1, rat INS-1 832/13, and mouse MIN6); two other cell types known to regulate glucose homeostasis and functionally utilize DOC2B (skeletal muscle rat myotube L6-GLUT4myc and human neuronal-like SH-SY5Y cells); and human islets sourced from individuals with no diabetes (ND). EVs derived from ND human plasma, ND human islets, and cell lines were isolated with either size exclusion chromatography or differential centrifugation. Isolated EVs were comprehensively characterized using dotblots, transmission electron microscopy, nanoparticle tracking analysis, and immunoblotting. Results DOC2B was present within EVs derived from ND human plasma, ND human islets, and INS-1 832/13 β-cells. Compared to neuronal-like SH-SY5Y cells and L6-GLUT4myc myotubes, clonal β-cells (EndoC-βH1, INS-1 832/13, and MIN6) produced significantly more EVs. DOC2B levels in EVs (over whole cell lysates) were higher in INS-1 832/13 β-cells compared to L6-GLUT4myc myotubes; SH-SY5Y neuronal-like cells did not release appreciable DOC2B. Mechanistically, we show that DOC2B was localized to the EV lumen; the tandem C2 domains were sufficient to confer sorting to INS-1 832/13 β-cell EVs. Discussion Clonal β-cells and ND human islets produce abundant EVs. In cell culture, appreciable DOC2B can be packaged into EVs, and a small fraction is excreted as a soluble protein. While DOC2B-laden EVs and soluble protein are present in ND plasma, further studies will be necessary to determine if DOC2B originating from β-cells significantly contributes to the plasma secretome.
Collapse
Affiliation(s)
- Diana Esparza
- Department of Molecular and Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute at City of Hope, Duarte, CA, United States
| | - Carinna Lima
- Department of Cancer Biology and Molecular Medicine, Beckman Research Institute at City of Hope, Duarte, CA, United States
| | - Sarah Abuelreich
- Department of Cancer Biology and Molecular Medicine, Beckman Research Institute at City of Hope, Duarte, CA, United States
| | - Ima Ghaeli
- Department of Cancer Biology and Molecular Medicine, Beckman Research Institute at City of Hope, Duarte, CA, United States
| | - Jinhee Hwang
- Department of Molecular and Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute at City of Hope, Duarte, CA, United States
| | - Eunjin Oh
- Department of Molecular and Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute at City of Hope, Duarte, CA, United States
| | - Ayelet Lenz
- Department of Molecular and Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute at City of Hope, Duarte, CA, United States
| | - Angel Gu
- Department of Translational Research and Cellular Therapeutics, Beckman Research Institute at City of Hope, Duarte, CA, United States
| | - Nan Jiang
- Department of Cancer Biology and Molecular Medicine, Beckman Research Institute at City of Hope, Duarte, CA, United States
| | - Fouad Kandeel
- Department of Translational Research and Cellular Therapeutics, Beckman Research Institute at City of Hope, Duarte, CA, United States
| | - Debbie C. Thurmond
- Department of Molecular and Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute at City of Hope, Duarte, CA, United States
| | - Tijana Jovanovic-Talisman
- Department of Cancer Biology and Molecular Medicine, Beckman Research Institute at City of Hope, Duarte, CA, United States
| |
Collapse
|
10
|
Sharma N, Angori S, Sandberg A, Mermelekas G, Lehtiö J, Wiklander OPB, Görgens A, Andaloussi SE, Eriksson H, Pernemalm M. Defining the Soluble and Extracellular Vesicle Protein Compartments of Plasma Using In-Depth Mass Spectrometry-Based Proteomics. J Proteome Res 2024; 23:4114-4127. [PMID: 39141927 PMCID: PMC11385381 DOI: 10.1021/acs.jproteome.4c00490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/16/2024]
Abstract
Plasma-derived extracellular vesicles (pEVs) are a potential source of diseased biomarker proteins. However, characterizing the pEV proteome is challenging due to its relatively low abundance and difficulties in enrichment. This study presents a streamlined workflow to identify EV proteins from cancer patient plasma using minimal sample input. Starting with 400 μL of plasma, we generated a comprehensive pEV proteome using size exclusion chromatography (SEC) combined with HiRIEF prefractionation-based mass spectrometry (MS). First, we compared the performance of HiRIEF and long gradient MS workflows using control pEVs, quantifying 2076 proteins with HiRIEF. In a proof-of-concept study, we applied SEC-HiRIEF-MS to a small cohort (12) of metastatic lung adenocarcinoma (LUAD) and malignant melanoma (MM) patients. We also analyzed plasma samples from the same patients to study the relationship between plasma and pEV proteomes. We identified and quantified 1583 proteins in cancer pEVs and 1468 proteins in plasma across all samples. While there was substantial overlap, the pEV proteome included several unique EV markers and cancer-related proteins. Differential analysis revealed 30 DEPs in LUAD vs the MM group, highlighting the potential of pEVs as biomarkers. This work demonstrates the utility of a prefractionation-based MS for comprehensive pEV proteomics and EV biomarker discovery. Data are available via ProteomeXchange with the identifiers PXD039338 and PXD038528.
Collapse
Affiliation(s)
- Nidhi Sharma
- Department of Oncology-Pathology, Karolinska Institute, 171 77 Stockholm, Sweden
- Science for Life Laboratory, Solna, Tomtebodavägen 23, 171 65 Solna, Sweden
| | - Silvia Angori
- Department of Oncology-Pathology, Karolinska Institute, 171 77 Stockholm, Sweden
| | - AnnSofi Sandberg
- Department of Oncology-Pathology, Karolinska Institute, 171 77 Stockholm, Sweden
- Science for Life Laboratory, Solna, Tomtebodavägen 23, 171 65 Solna, Sweden
| | - Georgios Mermelekas
- Department of Oncology-Pathology, Karolinska Institute, 171 77 Stockholm, Sweden
- Science for Life Laboratory, Solna, Tomtebodavägen 23, 171 65 Solna, Sweden
| | - Janne Lehtiö
- Department of Oncology-Pathology, Karolinska Institute, 171 77 Stockholm, Sweden
- Science for Life Laboratory, Solna, Tomtebodavägen 23, 171 65 Solna, Sweden
| | - Oscar P B Wiklander
- Theme Cancer, Skin Cancer Center, Karolinska University Hospital, 171 77 Solna, Sweden
- Biomolecular Medicine, Clinical Research Center, Department of Laboratory Medicine, Karolinska Institute, 171 76 Solna, Sweden
| | - André Görgens
- Biomolecular Medicine, Clinical Research Center, Department of Laboratory Medicine, Karolinska Institute, 171 76 Solna, Sweden
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, 45141 Essen, Germany
| | - Samir El Andaloussi
- Biomolecular Medicine, Clinical Research Center, Department of Laboratory Medicine, Karolinska Institute, 171 76 Solna, Sweden
| | - Hanna Eriksson
- Department of Oncology-Pathology, Karolinska Institute, 171 77 Stockholm, Sweden
- Theme Cancer, Skin Cancer Center, Karolinska University Hospital, 171 77 Solna, Sweden
- Science for Life Laboratory, Solna, Tomtebodavägen 23, 171 65 Solna, Sweden
| | - Maria Pernemalm
- Department of Oncology-Pathology, Karolinska Institute, 171 77 Stockholm, Sweden
- Science for Life Laboratory, Solna, Tomtebodavägen 23, 171 65 Solna, Sweden
| |
Collapse
|
11
|
Karpov OA, Stotland A, Raedschelders K, Chazarin B, Ai L, Murray CI, Van Eyk JE. Proteomics of the heart. Physiol Rev 2024; 104:931-982. [PMID: 38300522 PMCID: PMC11381016 DOI: 10.1152/physrev.00026.2023] [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: 07/03/2023] [Revised: 12/25/2023] [Accepted: 01/14/2024] [Indexed: 02/02/2024] Open
Abstract
Mass spectrometry-based proteomics is a sophisticated identification tool specializing in portraying protein dynamics at a molecular level. Proteomics provides biologists with a snapshot of context-dependent protein and proteoform expression, structural conformations, dynamic turnover, and protein-protein interactions. Cardiac proteomics can offer a broader and deeper understanding of the molecular mechanisms that underscore cardiovascular disease, and it is foundational to the development of future therapeutic interventions. This review encapsulates the evolution, current technologies, and future perspectives of proteomic-based mass spectrometry as it applies to the study of the heart. Key technological advancements have allowed researchers to study proteomes at a single-cell level and employ robot-assisted automation systems for enhanced sample preparation techniques, and the increase in fidelity of the mass spectrometers has allowed for the unambiguous identification of numerous dynamic posttranslational modifications. Animal models of cardiovascular disease, ranging from early animal experiments to current sophisticated models of heart failure with preserved ejection fraction, have provided the tools to study a challenging organ in the laboratory. Further technological development will pave the way for the implementation of proteomics even closer within the clinical setting, allowing not only scientists but also patients to benefit from an understanding of protein interplay as it relates to cardiac disease physiology.
Collapse
Affiliation(s)
- Oleg A Karpov
- Smidt Heart Institute, Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, United States
| | - Aleksandr Stotland
- Smidt Heart Institute, Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, United States
| | - Koen Raedschelders
- Smidt Heart Institute, Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, United States
| | - Blandine Chazarin
- Smidt Heart Institute, Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, United States
| | - Lizhuo Ai
- Smidt Heart Institute, Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, United States
| | - Christopher I Murray
- Smidt Heart Institute, Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, United States
| | - Jennifer E Van Eyk
- Smidt Heart Institute, Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, United States
| |
Collapse
|
12
|
Hu L, Zheng X, Zhou M, Wang J, Tong L, Dong M, Xu T, Li Z. Optimized AF4 combined with density cushion ultracentrifugation enables profiling of high-purity human blood extracellular vesicles. J Extracell Vesicles 2024; 13:e12470. [PMID: 39001700 PMCID: PMC11245685 DOI: 10.1002/jev2.12470] [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: 11/27/2023] [Accepted: 06/11/2024] [Indexed: 07/15/2024] Open
Abstract
Extracellular vesicles (EVs) have emerged as a promising tool for clinical liquid biopsy. However, the identification of EVs derived from blood samples is hindered by the presence of abundant plasma proteins, which impairs the downstream biochemical analysis of EV-associated proteins and nucleic acids. Here, we employed optimized asymmetric flow field-flow fractionation (AF4) combined with density cushion ultracentrifugation (UC) to obtain high-purity and intact EVs with very low lipoprotein contamination from human plasma and serum. Further proteomic analysis revealed more than 1000 EV-associated proteins, a large proportion of which has not been previously reported. Specifically, we found that cell-line-derived EV markers are incompatible with the identification of plasma-EVs and proposed that the proteins MYCT1, TSPAN14, MPIG6B and MYADM, as well as the traditional EV markers CD63 and CD147, are plasma-EV markers. Benefiting from the high-purity of EVs, we conducted comprehensive miRNA profiling of plasma EVs and nanosized particles (NPs), as well as compared plasma- and serum-derived EVs, which provides a valuable resource for the EV research community. Overall, our findings provide a comprehensive assessment of human blood EVs as a basis for clinical biopsy applications.
Collapse
Affiliation(s)
- Liqiao Hu
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and TechnologyHuazhong University of Science and TechnologyWuhanChina
- Guangzhou National LaboratoryGuangzhouChina
| | - Xinyue Zheng
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and TechnologyHuazhong University of Science and TechnologyWuhanChina
| | - Maoge Zhou
- Guangzhou National LaboratoryGuangzhouChina
| | - Jifeng Wang
- Laboratory of Proteomics, Institute of BiophysicsChinese Academy of SciencesBeijingChina
| | - Lingjun Tong
- Jinan Central Hospital, Medical Science and Technology Innovation CenterShandong First Medical University & Shandong Academy of Medical SciencesJinanChina
| | - Ming Dong
- Guangzhou National LaboratoryGuangzhouChina
| | - Tao Xu
- Guangzhou National LaboratoryGuangzhouChina
- Jinan Central Hospital, Medical Science and Technology Innovation CenterShandong First Medical University & Shandong Academy of Medical SciencesJinanChina
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of SciencesBeijingChina
- School of Biomedical EngineeringGuangzhou Medical UniversityGuangzhouChina
| | | |
Collapse
|
13
|
Espejo C, Ezenwa VO. Extracellular vesicles: an emerging tool for wild immunology. DISCOVERY IMMUNOLOGY 2024; 3:kyae011. [PMID: 39005930 PMCID: PMC11244269 DOI: 10.1093/discim/kyae011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 04/12/2024] [Accepted: 06/23/2024] [Indexed: 07/16/2024]
Abstract
The immune system is crucial for defending organisms against pathogens and maintaining health. Traditionally, research in immunology has relied on laboratory animals to understand how the immune system works. However, there is increasing recognition that wild animals, due to their greater genetic diversity, lifespan, and environmental exposures, have much to contribute to basic and translational immunology. Unfortunately, logistical challenges associated with collecting and storing samples from wildlife, and the lack of commercially available species-specific reagents have hindered the advancement of immunological research on wild species. Extracellular vesicles (EVs) are cell-derived nanoparticles present in all body fluids and tissues of organisms spanning from bacteria to mammals. Human and lab animal studies indicate that EVs are involved in a range of immunological processes, and recent work shows that EVs may play similar roles in diverse wildlife species. Thus, EVs can expand the toolbox available for wild immunology research, helping to overcome some of the challenges associated with this work. In this paper, we explore the potential application of EVs to wild immunology. First, we review current understanding of EV biology across diverse organisms. Next, we discuss key insights into the immune system gained from research on EVs in human and laboratory animal models and highlight emerging evidence from wild species. Finally, we identify research themes in wild immunology that can immediately benefit from the study of EVs and describe practical considerations for using EVs in wildlife research.
Collapse
Affiliation(s)
- Camila Espejo
- Department of Ecology and Evolutionary Biology, Yale University, New Haven, CT, USA
| | - Vanessa O Ezenwa
- Department of Ecology and Evolutionary Biology, Yale University, New Haven, CT, USA
| |
Collapse
|
14
|
Enomoto T, Shirai Y, Takeda Y, Edahiro R, Shichino S, Nakayama M, Takahashi-Itoh M, Noda Y, Adachi Y, Kawasaki T, Koba T, Futami Y, Yaga M, Hosono Y, Yoshimura H, Amiya S, Hara R, Yamamoto M, Nakatsubo D, Suga Y, Naito M, Masuhiro K, Hirata H, Iwahori K, Nagatomo I, Miyake K, Koyama S, Fukushima K, Shiroyama T, Naito Y, Futami S, Natsume-Kitatani Y, Nojima S, Yanagawa M, Shintani Y, Nogami-Itoh M, Mizuguchi K, Adachi J, Tomonaga T, Inoue Y, Kumanogoh A. SFTPB in serum extracellular vesicles as a biomarker of progressive pulmonary fibrosis. JCI Insight 2024; 9:e177937. [PMID: 38855869 PMCID: PMC11382876 DOI: 10.1172/jci.insight.177937] [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: 11/28/2023] [Accepted: 04/23/2024] [Indexed: 06/11/2024] Open
Abstract
Progressive pulmonary fibrosis (PPF), defined as the worsening of various interstitial lung diseases (ILDs), currently lacks useful biomarkers. To identify novel biomarkers for early detection of patients at risk of PPF, we performed a proteomic analysis of serum extracellular vesicles (EVs). Notably, the identified candidate biomarkers were enriched for lung-derived proteins participating in fibrosis-related pathways. Among them, pulmonary surfactant-associated protein B (SFTPB) in serum EVs could predict ILD progression better than the known biomarkers, serum KL-6 and SP-D, and it was identified as an independent prognostic factor from ILD-gender-age-physiology index. Subsequently, the utility of SFTPB for predicting ILD progression was evaluated further in 2 cohorts using serum EVs and serum, respectively, suggesting that SFTPB in serum EVs but not in serum was helpful. Among SFTPB forms, pro-SFTPB levels were increased in both serum EVs and lungs of patients with PPF compared with those of the control. Consistently, in a mouse model, the levels of pro-SFTPB, primarily originating from alveolar epithelial type 2 cells, were increased similarly in serum EVs and lungs, reflecting pro-fibrotic changes in the lungs, as supported by single-cell RNA sequencing. SFTPB, especially its pro-form, in serum EVs could serve as a biomarker for predicting ILD progression.
Collapse
Affiliation(s)
| | - Yuya Shirai
- Department of Respiratory Medicine and Clinical Immunology and
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Yoshito Takeda
- Department of Respiratory Medicine and Clinical Immunology and
| | - Ryuya Edahiro
- Department of Respiratory Medicine and Clinical Immunology and
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Shigeyuki Shichino
- Division of Molecular Regulation of Inflammatory and Immune Diseases, Research Institute of Biomedical Sciences, Tokyo University of Science, Chiba, Japan
| | - Mana Nakayama
- Department of Respiratory Medicine and Clinical Immunology and
| | | | - Yoshimi Noda
- Department of Respiratory Medicine and Clinical Immunology and
| | - Yuichi Adachi
- Department of Respiratory Medicine and Clinical Immunology and
| | | | - Taro Koba
- Department of Respiratory Medicine and Clinical Immunology and
| | - Yu Futami
- Department of Respiratory Medicine and Clinical Immunology and
- Department of Respiratory Medicine, Kinki Central Hospital of the Mutual Aid Association of Public School Teachers, Itami, Hyogo, Japan
| | - Moto Yaga
- Department of Respiratory Medicine and Clinical Immunology and
| | - Yuki Hosono
- Department of Respiratory Medicine and Clinical Immunology and
| | | | - Saori Amiya
- Department of Respiratory Medicine and Clinical Immunology and
| | - Reina Hara
- Department of Respiratory Medicine and Clinical Immunology and
| | - Makoto Yamamoto
- Department of Respiratory Medicine and Clinical Immunology and
| | | | - Yasuhiko Suga
- Department of Respiratory Medicine and Clinical Immunology and
| | - Maiko Naito
- Department of Respiratory Medicine and Clinical Immunology and
| | | | - Haruhiko Hirata
- Department of Respiratory Medicine and Clinical Immunology and
| | - Kota Iwahori
- Department of Respiratory Medicine and Clinical Immunology and
| | - Izumi Nagatomo
- Department of Respiratory Medicine and Clinical Immunology and
| | - Kotaro Miyake
- Department of Respiratory Medicine and Clinical Immunology and
| | - Shohei Koyama
- Department of Respiratory Medicine and Clinical Immunology and
| | | | | | - Yujiro Naito
- Department of Respiratory Medicine and Clinical Immunology and
| | - Shinji Futami
- Department of Respiratory Medicine and Clinical Immunology and
| | - Yayoi Natsume-Kitatani
- Laboratory of Bioinformatics, Artificial Intelligence Center for Health and Biomedical Research, National Institutes of Biomedical Innovation, Health and Nutrition, Settsu, Osaka, Japan
- Institute of Advanced Medical Sciences, Tokushima University, Tokushima, Japan
| | | | | | - Yasushi Shintani
- Department of General Thoracic Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Mari Nogami-Itoh
- Laboratory of Bioinformatics, Artificial Intelligence Center for Health and Biomedical Research, National Institutes of Biomedical Innovation, Health and Nutrition, Settsu, Osaka, Japan
| | - Kenji Mizuguchi
- Laboratory of Bioinformatics, Artificial Intelligence Center for Health and Biomedical Research, National Institutes of Biomedical Innovation, Health and Nutrition, Settsu, Osaka, Japan
- Laboratory for Computational Biology, Institute for Protein Research, Osaka University, Suita, Osaka, Japan
| | - Jun Adachi
- Laboratory of Proteomics for Drug Discovery, Center for Drug Design Research, National Institutes of Biomedical Innovation, Health and Nutrition, Ibaraki, Osaka, Japan
| | - Takeshi Tomonaga
- Laboratory of Proteomics for Drug Discovery, Center for Drug Design Research, National Institutes of Biomedical Innovation, Health and Nutrition, Ibaraki, Osaka, Japan
- Proteobiologics Co., Ltd., Minoh, Osaka, Japan
| | - Yoshikazu Inoue
- Clinical Research Center, NHO Kinki Chuo Chest Medical Center, Sakai, Osaka, Japan
- Osaka Anti-tuberculosis Association, Osaka Fukujuji Hospital, Neyagawa, Osaka, Japan
| | - Atsushi Kumanogoh
- Department of Respiratory Medicine and Clinical Immunology and
- Center for Infectious Diseases for Education and Research (CiDER)
- Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives (OTRI)
- Department of Immunopathology, Immunology Frontier Research Center (WPI-IFReC); and
- Japan Agency for Medical Research and Development-Core Research for Evolutional Science and Technology (AMED-CREST), Osaka University, Suita, Osaka, Japan
| |
Collapse
|
15
|
Sakane S, Hikita H, Shirai K, Sakamoto T, Narumi R, Adachi J, Kakita N, Yamada Y, Toyoda H, Takahashi H, Suda G, Kai M, Tahata Y, Sakamori R, Kumazaki S, Fukumoto K, Myojin Y, Murai K, Kodama T, Tatsumi T, Tomonaga T, Sakamoto N, Morii E, Takehara T. Proteomic analysis of serum extracellular vesicles reveals Fibulin-3 as a new marker predicting liver-related events in MASLD. Hepatol Commun 2024; 8:e0448. [PMID: 38829196 PMCID: PMC11150025 DOI: 10.1097/hc9.0000000000000448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Accepted: 03/05/2024] [Indexed: 06/05/2024] Open
Abstract
BACKGROUND There is a need for novel noninvasive markers for metabolic dysfunction-associated steatotic liver disease (MASLD) to stratify patients at high risk for liver-related events including liver cancer and decompensation. In the present study, we used proteomic analysis of proteins in extracellular vesicles (EVs) to identify new biomarkers that change with fibrosis progression and can predict the development of liver-related events. METHODS We analyzed serum EVs from 50 patients with MASLD assessed for liver fibrosis by biopsy and identified proteins that altered with advanced fibrosis. A further evaluation was conducted on another cohort of 463 patients with MASLD with biopsy. RESULTS Eight candidate proteins were identified by proteomic analysis of serum EVs. Among them, serum levels of Fibulin-3, Fibulin-1, and Ficolin 1 correlated with their EV levels. In addition, serum Fibulin-3 and serum Fibulin-1 levels changed significantly with advanced fibrosis. Using another cohort with biopsy, we found that the serum Fibulin-3 concentration was significantly greater in those with advanced fibrosis but that the serum Fibulin-1 concentration was not significantly different. Multivariate Cox proportional hazards analysis revealed that a higher Fibrosis-4 (FIB-4) index and higher serum Fibulin-3 concentration were independent risk factors for liver-related events. When the cutoff value for the serum Fibulin-3 concentration was 6.0 µg/mL according to the Youden index of AUROCs, patients with high serum Fibulin-3 significantly more frequently developed liver-related events than did other patients. Validation using another cohort of 226 patients with clinically diagnosed MASLD confirmed that high serum Fibulin-3 levels are associated with a greater frequency of liver-related events. CONCLUSIONS Serum Fibulin-3 was identified as a biomarker for predicting liver-related events in patients with MASLD.
Collapse
Affiliation(s)
- Sadatsugu Sakane
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Hayato Hikita
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Kumiko Shirai
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Tatsuya Sakamoto
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Ryohei Narumi
- Laboratory of Proteomics for Drug Discovery, Center for Drug Design Research, National Institute of Biomedical Innovation, Health and Nutrition, Osaka, Japan
| | - Jun Adachi
- Laboratory of Proteomics for Drug Discovery, Center for Drug Design Research, National Institute of Biomedical Innovation, Health and Nutrition, Osaka, Japan
| | - Naruyasu Kakita
- Department of Gastroenterology and Hepatology, Kaizuka City Hospital, Osaka, Japan
| | - Yukinori Yamada
- Department of Gastroenterology and Hepatology, Kaizuka City Hospital, Osaka, Japan
| | - Hidenori Toyoda
- Department of Gastroenterology, Ogaki Municipal Hospital, Ogaki, Japan
| | | | - Goki Suda
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Machiko Kai
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Yuki Tahata
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Ryotaro Sakamori
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Shusuke Kumazaki
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Kenji Fukumoto
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Yuta Myojin
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Kazuhiro Murai
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Takahiro Kodama
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Tomohide Tatsumi
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Takeshi Tomonaga
- Laboratory of Proteomics for Drug Discovery, Center for Drug Design Research, National Institute of Biomedical Innovation, Health and Nutrition, Osaka, Japan
| | - Naoya Sakamoto
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Eiichi Morii
- Department of Pathology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Tetsuo Takehara
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Osaka University, Osaka, Japan
| |
Collapse
|
16
|
Maeda Y, Watanabe Y, Ishikawa N, Yoshida T, Kimura N, Abe H, Sakamaki A, Kamimura H, Yokoo T, Kamimura K, Tsuchiya A, Terai S. Platelet-rich plasma-derived extracellular vesicles improve liver cirrhosis in mice. Regen Ther 2024; 26:1048-1057. [PMID: 39569343 PMCID: PMC11576940 DOI: 10.1016/j.reth.2024.10.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Revised: 10/18/2024] [Accepted: 10/23/2024] [Indexed: 11/22/2024] Open
Abstract
INTRODUCTION Cirrhosis remains a significant clinical challenge due to its poor prognosis and limited treatment options, creating a high unmet medical need for the development of novel therapies. In this study, we analyzed the effects of a novel approach to treat cirrhosis using platelet-rich plasma-derived extracellular vesicles (PRPEV) in mice. METHODS PRPEV were collected from platelet-rich plasma using ultrafiltration, and their proteomes were analyzed. The carbon tetrachloride (CCl4)-induced cirrhosis model of mice was used to evaluate the effect of PRPEV administration and compared with the control group (n = 8). In vitro and in vivo mechanistic analyses of PRPEV administration were confirmed using real time-PCR and immunostaining. RESULTS Gene ontology analysis based on the proteome revealed that PRPEV contain many factors associated with EV and immune responses. In vitro, PRPEV polarize macrophages into an anti-inflammatory phenotype. Following PRPEV administration, there was a decrease in serum alanine aminotransferase levels and reduction in liver fibrosis, while mRNA levels of regenerative factors were upregulated and transforming growth factor β-1 was downregulated. Furthermore, the number of anti-inflammatory macrophages in the liver increased. CONCLUSIONS PRPEV may contribute to hepatocyte proliferation, anti-inflammation, and anti-fibrogenesis in the liver. This novel concept paves the way for cirrhosis treatment.
Collapse
Affiliation(s)
- Yuichirou Maeda
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Yusuke Watanabe
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
- Division of Preemptive Medicine for Digestive Disease and Healthy Active Life, School of Medicine, Niigata University, Niigata, Japan
| | - Natsuki Ishikawa
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Tomoaki Yoshida
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
- Division of Preemptive Medicine for Digestive Disease and Healthy Active Life, School of Medicine, Niigata University, Niigata, Japan
| | - Naruhiro Kimura
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Hiroyuki Abe
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Akira Sakamaki
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Hiroteru Kamimura
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Takeshi Yokoo
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
- Division of Preemptive Medicine for Digestive Disease and Healthy Active Life, School of Medicine, Niigata University, Niigata, Japan
| | - Kenya Kamimura
- Department of General Medicine, Niigata University School of Medicine, Niigata University, Niigata, Japan
| | - Atsunori Tsuchiya
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
- Future Medical Research Center for Exosome and Designer Cells (F-EDC), Niigata University, Japan
| | - Shuji Terai
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
- Future Medical Research Center for Exosome and Designer Cells (F-EDC), Niigata University, Japan
| |
Collapse
|
17
|
Kistenmacher S, Schwämmle M, Martin G, Ulrich E, Tholen S, Schilling O, Gießl A, Schlötzer-Schrehardt U, Bucher F, Schlunck G, Nazarenko I, Reinhard T, Polisetti N. Enrichment, Characterization, and Proteomic Profiling of Small Extracellular Vesicles Derived from Human Limbal Mesenchymal Stromal Cells and Melanocytes. Cells 2024; 13:623. [PMID: 38607062 PMCID: PMC11011788 DOI: 10.3390/cells13070623] [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: 03/11/2024] [Revised: 03/27/2024] [Accepted: 03/28/2024] [Indexed: 04/13/2024] Open
Abstract
Limbal epithelial progenitor cells (LEPC) rely on their niche environment for proper functionality and self-renewal. While extracellular vesicles (EV), specifically small EVs (sEV), have been proposed to support LEPC homeostasis, data on sEV derived from limbal niche cells like limbal mesenchymal stromal cells (LMSC) remain limited, and there are no studies on sEVs from limbal melanocytes (LM). In this study, we isolated sEV from conditioned media of LMSC and LM using a combination of tangential flow filtration and size exclusion chromatography and characterized them by nanoparticle tracking analysis, transmission electron microscopy, Western blot, multiplex bead arrays, and quantitative mass spectrometry. The internalization of sEV by LEPC was studied using flow cytometry and confocal microscopy. The isolated sEVs exhibited typical EV characteristics, including cell-specific markers such as CD90 for LMSC-sEV and Melan-A for LM-sEV. Bioinformatics analysis of the proteomic data suggested a significant role of sEVs in extracellular matrix deposition, with LMSC-derived sEV containing proteins involved in collagen remodeling and cell matrix adhesion, whereas LM-sEV proteins were implicated in other cellular bioprocesses such as cellular pigmentation and development. Moreover, fluorescently labeled LMSC-sEV and LM-sEV were taken up by LEPC and localized to their perinuclear compartment. These findings provide valuable insights into the complex role of sEV from niche cells in regulating the human limbal stem cell niche.
Collapse
Affiliation(s)
- Sebastian Kistenmacher
- Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, Killianstrasse 5, 79106 Freiburg, Germany
| | - Melanie Schwämmle
- Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, Killianstrasse 5, 79106 Freiburg, Germany
- Faculty of Biology, University of Freiburg, Schaenzlestrasse 1, D–79104 Freiburg, Germany
| | - Gottfried Martin
- Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, Killianstrasse 5, 79106 Freiburg, Germany
| | - Eva Ulrich
- Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, Killianstrasse 5, 79106 Freiburg, Germany
| | - Stefan Tholen
- Institute of Surgical Pathology, Faculty of Medicine, Freiburg, Medical Center, University of Freiburg, 79085 Freiburg im Breisgau, Germany
| | - Oliver Schilling
- Institute of Surgical Pathology, Faculty of Medicine, Freiburg, Medical Center, University of Freiburg, 79085 Freiburg im Breisgau, Germany
| | - Andreas Gießl
- Department of Ophthalmology, University Hospital Erlangen, Friedrich-Alexander-University of Erlan-gen-Nürnberg, Schwabachanlage 6, 91054 Erlangen, Germany
| | - Ursula Schlötzer-Schrehardt
- Department of Ophthalmology, University Hospital Erlangen, Friedrich-Alexander-University of Erlan-gen-Nürnberg, Schwabachanlage 6, 91054 Erlangen, Germany
| | - Felicitas Bucher
- Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, Killianstrasse 5, 79106 Freiburg, Germany
| | - Günther Schlunck
- Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, Killianstrasse 5, 79106 Freiburg, Germany
| | - Irina Nazarenko
- Institute for Infection Prevention and Hospital Epidemiology, Faculty of Medicine, Medical Center, University of Freiburg, 79106 Freiburg, Germany
| | - Thomas Reinhard
- Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, Killianstrasse 5, 79106 Freiburg, Germany
| | - Naresh Polisetti
- Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, Killianstrasse 5, 79106 Freiburg, Germany
| |
Collapse
|
18
|
Arredondo-Damián JG, Martínez-Soto JM, Molina-Pelayo FA, Soto-Guzmán JA, Castro-Sánchez L, López-Soto LF, Candia-Plata MDC. Systematic review and bioinformatics analysis of plasma and serum extracellular vesicles proteome in type 2 diabetes. Heliyon 2024; 10:e25537. [PMID: 38356516 PMCID: PMC10865249 DOI: 10.1016/j.heliyon.2024.e25537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 01/28/2024] [Accepted: 01/29/2024] [Indexed: 02/16/2024] Open
Abstract
Background Type 2 diabetes (T2D) is a complex metabolic ailment marked by a global high prevalence and significant attention in primary healthcare settings due to its elevated morbidity and mortality rates. The pathophysiological mechanisms underlying the onset and progression of this disease remain subjects of ongoing investigation. Recent evidence underscores the pivotal role of the intricate intercellular communication network, wherein cell-derived vesicles, commonly referred to as extracellular vesicles (EVs), emerge as dynamic regulators of diabetes-related complications. Given that the protein cargo carried by EVs is contingent upon the metabolic conditions of the originating cells, particular proteins may serve as informative indicators for the risk of activating or inhibiting signaling pathways crucial to the progression of T2D complications. Methods In this study, we conducted a systematic review to analyze the published evidence on the proteome of EVs from the plasma or serum of patients with T2D, both with and without complications (PROSPERO: CRD42023431464). Results Nine eligible articles were systematically identified from the databases, and the proteins featured in these articles underwent Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis. We identified changes in the level of 426 proteins, with CST6, CD55, HBA1, S100A8, and S100A9 reported to have high levels, while FGL1 exhibited low levels. Conclusion These proteins are implicated in pathophysiological mechanisms such as inflammation, complement, and platelet activation, suggesting their potential as risk markers for T2D development and progression. Further studies are required to explore this topic in greater detail.
Collapse
Affiliation(s)
| | | | | | | | - Luis Castro-Sánchez
- University Center for Biomedical Research, University of Colima, Colima, Colima, Mexico
- CONAHCYT-University of Colima, Colima, Colima, Mexico
| | | | | |
Collapse
|
19
|
Lattmann E, Räss L, Tognetti M, Gómez JMM, Lapaire V, Bruderer R, Reiter L, Feng Y, Steinmetz LM, Levesque MP. Size-exclusion chromatography combined with DIA-MS enables deep proteome profiling of extracellular vesicles from melanoma plasma and serum. Cell Mol Life Sci 2024; 81:90. [PMID: 38353833 PMCID: PMC10867102 DOI: 10.1007/s00018-024-05137-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: 04/23/2023] [Revised: 01/18/2024] [Accepted: 01/19/2024] [Indexed: 02/16/2024]
Abstract
Extracellular vesicles (EVs) are important players in melanoma progression, but their use as clinical biomarkers has been limited by the difficulty of profiling blood-derived EV proteins with high depth of coverage, the requirement for large input amounts, and complex protocols. Here, we provide a streamlined and reproducible experimental workflow to identify plasma- and serum- derived EV proteins of healthy donors and melanoma patients using minimal amounts of sample input. SEC-DIA-MS couples size-exclusion chromatography to EV concentration and deep-proteomic profiling using data-independent acquisition. From as little as 200 µL of plasma per patient in a cohort of three healthy donors and six melanoma patients, we identified and quantified 2896 EV-associated proteins, achieving a 3.5-fold increase in depth compared to previously published melanoma studies. To compare the EV-proteome to unenriched blood, we employed an automated workflow to deplete the 14 most abundant proteins from plasma and serum and thereby approximately doubled protein group identifications versus native blood. The EV proteome diverged from corresponding unenriched plasma and serum, and unlike the latter, separated healthy donor and melanoma patient samples. Furthermore, known melanoma markers, such as MCAM, TNC, and TGFBI, were upregulated in melanoma EVs but not in depleted melanoma plasma, highlighting the specific information contained in EVs. Overall, EVs were significantly enriched in intact membrane proteins and proteins related to SNARE protein interactions and T-cell biology. Taken together, we demonstrated the increased sensitivity of an EV-based proteomic workflow that can be easily applied to larger melanoma cohorts and other indications.
Collapse
Affiliation(s)
- Evelyn Lattmann
- Department of Dermatology, University Hospital Zurich, University of Zurich, Schlieren, Switzerland
| | - Luca Räss
- Biognosys AG, Schlieren, Switzerland
| | | | - Julia M Martínez Gómez
- Department of Dermatology, University Hospital Zurich, University of Zurich, Schlieren, Switzerland
| | - Valérie Lapaire
- Department of Dermatology, University Hospital Zurich, University of Zurich, Schlieren, Switzerland
| | | | | | | | - Lars M Steinmetz
- Stanford Genome Technology Center, Stanford University, Palo Alto, CA, USA.
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA.
- Genome Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany.
| | - Mitchell P Levesque
- Department of Dermatology, University Hospital Zurich, University of Zurich, Schlieren, Switzerland.
| |
Collapse
|
20
|
Yuyama K, Sun H, Fujii R, Hemmi I, Ueda K, Igeta Y. Extracellular vesicle proteome unveils cathepsin B connection to Alzheimer's disease pathogenesis. Brain 2024; 147:627-636. [PMID: 38071653 PMCID: PMC10834236 DOI: 10.1093/brain/awad361] [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: 04/26/2023] [Revised: 09/27/2023] [Accepted: 10/10/2023] [Indexed: 02/03/2024] Open
Abstract
Extracellular vesicles (EVs) are membrane vesicles that are released extracellularly and considered to be implicated in the pathogenesis of neurodegenerative diseases including Alzheimer's disease. Here, CSF EVs of 16 ATN-classified cases were subjected to quantitative proteome analysis. In these CSF EVs, levels of 11 proteins were significantly altered during the ATN stage transitions (P < 0.05 and fold-change > 2.0). These proteins were thought to be associated with Alzheimer's disease pathogenesis and represent candidate biomarkers for pathogenic stage classification. Enzyme-linked immunosorbent assay analysis of CSF and plasma EVs revealed altered levels of cathepsin B (CatB) during the ATN transition (seven ATN groups in validation set, n = 136). The CSF and plasma EV CatB levels showed a negative correlation with CSF amyloid-β42 concentrations. This proteomic landscape of CSF EVs in ATN classifications can depict the molecular framework of Alzheimer's disease progression, and CatB may be considered a promising candidate biomarker and therapeutic target in Alzheimer's disease amyloid pathology.
Collapse
Affiliation(s)
- Kohei Yuyama
- Lipid Biofunction Section, Faculty of Advanced Life Science, Hokkaido University, Sapporo 001-0021, Japan
| | - Hui Sun
- Lipid Biofunction Section, Faculty of Advanced Life Science, Hokkaido University, Sapporo 001-0021, Japan
| | - Risa Fujii
- Cancer Proteomics Group, Cancer Precision Medicine Center, Japanese Foundation for Cancer Research, Tokyo 035-8550, Japan
| | - Isao Hemmi
- Department of Nursing, Japanese Red Cross College of Nursing, Tokyo 150-0012, Japan
| | - Koji Ueda
- Cancer Proteomics Group, Cancer Precision Medicine Center, Japanese Foundation for Cancer Research, Tokyo 035-8550, Japan
| | - Yukifusa Igeta
- Department of Dementia, Dementia Center, Toranomon Hospital, Tokyo 105-8470, Japan
- Division of Dementia Research, Okinaka Memorial Institute for Medical Research, Tokyo 105-8470, Japan
| |
Collapse
|
21
|
Nakajima D, Konno R, Miyashita Y, Ishikawa M, Ohara O, Kawashima Y. Proteome Analysis of Serum Purified Using Solanum tuberosum and Lycopersicon esculentum Lectins. Int J Mol Sci 2024; 25:1315. [PMID: 38279312 PMCID: PMC10816257 DOI: 10.3390/ijms25021315] [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: 12/27/2023] [Revised: 01/17/2024] [Accepted: 01/19/2024] [Indexed: 01/28/2024] Open
Abstract
Serum and plasma exhibit a broad dynamic range of protein concentrations, posing challenges for proteome analysis. Various technologies have been developed to reduce this complexity, including high-abundance depletion methods utilizing antibody columns, extracellular vesicle enrichment techniques, and trace protein enrichment using nanobead cocktails. Here, we employed lectins to address this, thereby extending the scope of biomarker discovery in serum or plasma using a novel approach. We enriched serum proteins using 37 different lectins and subjected them to LC-MS/MS analysis with data-independent acquisition. Solanum tuberosum lectin (STL) and Lycopersicon esculentum lectin (LEL) enabled the detection of more serum proteins than the other lectins. STL and LEL bind to N-acetylglucosamine oligomers, emphasizing the significance of capturing these oligomer-binding proteins when analyzing serum trace proteins. Combining STL and LEL proved more effective than using them separately, allowing us to identify over 3000 proteins from serum through single-shot proteome analysis. We applied the STL/LEL trace-protein enrichment method to the sera of systemic lupus erythematosus model mice. This revealed differences in >1300 proteins between the systemic lupus erythematosus model and control mouse sera, underscoring the utility of this method for biomarker discovery.
Collapse
Affiliation(s)
- Daisuke Nakajima
- Department of Applied Genomics, Kazusa DNA Research Institute, 2-5-23 Kazusa Kamatari, Kisarazu 292-0818, Chiba, Japan; (D.N.); (R.K.); (Y.M.); (M.I.); (O.O.)
| | - Ryo Konno
- Department of Applied Genomics, Kazusa DNA Research Institute, 2-5-23 Kazusa Kamatari, Kisarazu 292-0818, Chiba, Japan; (D.N.); (R.K.); (Y.M.); (M.I.); (O.O.)
| | - Yasuomi Miyashita
- Department of Applied Genomics, Kazusa DNA Research Institute, 2-5-23 Kazusa Kamatari, Kisarazu 292-0818, Chiba, Japan; (D.N.); (R.K.); (Y.M.); (M.I.); (O.O.)
- Department of Developmental Biology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Chiba, Japan
| | - Masaki Ishikawa
- Department of Applied Genomics, Kazusa DNA Research Institute, 2-5-23 Kazusa Kamatari, Kisarazu 292-0818, Chiba, Japan; (D.N.); (R.K.); (Y.M.); (M.I.); (O.O.)
| | - Osamu Ohara
- Department of Applied Genomics, Kazusa DNA Research Institute, 2-5-23 Kazusa Kamatari, Kisarazu 292-0818, Chiba, Japan; (D.N.); (R.K.); (Y.M.); (M.I.); (O.O.)
| | - Yusuke Kawashima
- Department of Applied Genomics, Kazusa DNA Research Institute, 2-5-23 Kazusa Kamatari, Kisarazu 292-0818, Chiba, Japan; (D.N.); (R.K.); (Y.M.); (M.I.); (O.O.)
| |
Collapse
|
22
|
Tian W, Shi D, Zhang Y, Wang H, Tang H, Han Z, Wong CCL, Cui L, Zheng J, Chen Y. Deep proteomic analysis of obstetric antiphospholipid syndrome by DIA-MS of extracellular vesicle enriched fractions. Commun Biol 2024; 7:99. [PMID: 38225453 PMCID: PMC10789860 DOI: 10.1038/s42003-024-05789-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 01/03/2024] [Indexed: 01/17/2024] Open
Abstract
Proteins in the plasma/serum mirror an individual's physiology. Circulating extracellular vesicles (EVs) proteins constitute a large portion of the plasma/serum proteome. Thus, deep and unbiased proteomic analysis of circulating plasma/serum extracellular vesicles holds promise for discovering disease biomarkers as well as revealing disease mechanisms. We established a workflow for simple, deep, and reproducible proteome analysis of both serum large and small EVs enriched fractions by ultracentrifugation plus 4D-data-independent acquisition mass spectrometry (4D-DIA-MS). In our cohort study of obstetric antiphospholipid syndrome (OAPS), 4270 and 3328 proteins were identified from large and small EVs enriched fractions respectively. Both of them revealed known or new pathways related to OAPS. Increased levels of von Willebrand factor (VWF) and insulin receptor (INSR) were identified as candidate biomarkers, which shed light on hypercoagulability and abnormal insulin signaling in disease progression. Our workflow will significantly promote our understanding of plasma/serum-based disease mechanisms and generate new biomarkers.
Collapse
Affiliation(s)
- Wenmin Tian
- Department of Biochemistry and Biophysics, Center for Precision Medicine Multi-Omics Research, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Dongxue Shi
- Department of Biochemistry and Biophysics, Center for Precision Medicine Multi-Omics Research, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Yinmei Zhang
- Department of Laboratory Medicine, Peking University Third Hospital, Beijing, P R China
| | - Hongli Wang
- Department of Biochemistry and Biophysics, Center for Precision Medicine Multi-Omics Research, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Haohao Tang
- Department of Biochemistry and Biophysics, Center for Precision Medicine Multi-Omics Research, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Zhongyu Han
- Department of Laboratory Medicine, Peking University Third Hospital, Beijing, P R China
| | - Catherine C L Wong
- Department of Medical Research Center, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, 100730, Beijing, China
- Tsinghua University-Peking University Joint Center for Life Sciences, Peking University, 100084, Beijing, China
| | - Liyan Cui
- Department of Laboratory Medicine, Peking University Third Hospital, Beijing, P R China.
| | - Jiajia Zheng
- Department of Laboratory Medicine, Peking University Third Hospital, Beijing, P R China.
| | - Yang Chen
- Department of Biochemistry and Biophysics, Center for Precision Medicine Multi-Omics Research, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China.
| |
Collapse
|
23
|
Tomoto M, Mineharu Y, Sato N, Tamada Y, Nogami-Itoh M, Kuroda M, Adachi J, Takeda Y, Mizuguchi K, Kumanogoh A, Natsume-Kitatani Y, Okuno Y. Idiopathic pulmonary fibrosis-specific Bayesian network integrating extracellular vesicle proteome and clinical information. Sci Rep 2024; 14:1315. [PMID: 38225283 PMCID: PMC10789725 DOI: 10.1038/s41598-023-50905-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 12/27/2023] [Indexed: 01/17/2024] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive disease characterized by severe lung fibrosis and a poor prognosis. Although the biomolecules related to IPF have been extensively studied, molecular mechanisms of the pathogenesis and their association with serum biomarkers and clinical findings have not been fully elucidated. We constructed a Bayesian network using multimodal data consisting of a proteome dataset from serum extracellular vesicles, laboratory examinations, and clinical findings from 206 patients with IPF and 36 controls. Differential protein expression analysis was also performed by edgeR and incorporated into the constructed network. We have successfully visualized the relationship between biomolecules and clinical findings with this approach. The IPF-specific network included modules associated with TGF-β signaling (TGFB1 and LRC32), fibrosis-related (A2MG and PZP), myofibroblast and inflammation (LRP1 and ITIH4), complement-related (SAA1 and SAA2), as well as serum markers, and clinical symptoms (KL-6, SP-D and fine crackles). Notably, it identified SAA2 associated with lymphocyte counts and PSPB connected with the serum markers KL-6 and SP-D, along with fine crackles as clinical manifestations. These results contribute to the elucidation of the pathogenesis of IPF and potential therapeutic targets.
Collapse
Affiliation(s)
- Mei Tomoto
- Department of Biomedical Data Intelligence, Kyoto University Graduate School of Medicine, 54 Shogoin Kawahara-Cho, Sakyo-Ku, Kyoto, 606-8507, Japan
| | - Yohei Mineharu
- Department of Biomedical Data Intelligence, Kyoto University Graduate School of Medicine, 54 Shogoin Kawahara-Cho, Sakyo-Ku, Kyoto, 606-8507, Japan
- Department of Artificial Intelligence in Healthcare and Medicine, Kyoto University Graduate School of Medicine, 54 Shogoin Kawahara-Cho, Sakyo-Ku, Kyoto, 606-8507, Japan
| | - Noriaki Sato
- Department of Biomedical Data Intelligence, Kyoto University Graduate School of Medicine, 54 Shogoin Kawahara-Cho, Sakyo-Ku, Kyoto, 606-8507, Japan
- Human Genome Center, The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokane-Dai, Minato-Ku, Tokyo, 108-8639, Japan
| | - Yoshinori Tamada
- Innovation Center for Health Promotion, Hirosaki University, 5 Zaifu-Cho Hirosaki City, Aomori, 036-8562, Japan
| | - Mari Nogami-Itoh
- Artificial Intelligence Center for Health and Biomedical Research, National Institutes of Biomedical Innovation, Health and Nutrition, 3-17, Senrioka-Shinmachi, Settsu City, Osaka, 566-0002, Japan
| | - Masataka Kuroda
- Artificial Intelligence Center for Health and Biomedical Research, National Institutes of Biomedical Innovation, Health and Nutrition, 3-17, Senrioka-Shinmachi, Settsu City, Osaka, 566-0002, Japan
- Discovery Technology Laboratories, Mitsubishi Tanabe Pharma Corporation, 1000, Kamoshida-cho, Aoba-ku, Yokohama, Kanagawa, 227-0033, Japan
| | - Jun Adachi
- Laboratory of Proteomics for Drug Discovery, Center for Drug Design Research, National Institutes of Biomedical Innovation, Health and Nutrition, 7-6-8 Saito-Asagi, Ibaraki, Osaka, 567-0085, Japan
| | - Yoshito Takeda
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, 2-2 Yamada-Oka, Suita City, Osaka, 565-0871, Japan
| | - Kenji Mizuguchi
- Artificial Intelligence Center for Health and Biomedical Research, National Institutes of Biomedical Innovation, Health and Nutrition, 3-17, Senrioka-Shinmachi, Settsu City, Osaka, 566-0002, Japan
- Institute for Protein Research, Osaka University, 3-2 Yamada-Oka, Suita City, Osaka, 565-0871, Japan
| | - Atsushi Kumanogoh
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, 2-2 Yamada-Oka, Suita City, Osaka, 565-0871, Japan
| | - Yayoi Natsume-Kitatani
- Innovation Center for Health Promotion, Hirosaki University, 5 Zaifu-Cho Hirosaki City, Aomori, 036-8562, Japan.
- Artificial Intelligence Center for Health and Biomedical Research, National Institutes of Biomedical Innovation, Health and Nutrition, 3-17, Senrioka-Shinmachi, Settsu City, Osaka, 566-0002, Japan.
- Institute of Advanced Medical Sciences, Tokushima University, 3-18-15, Kuramoto-Cho, Tokushima City, Tokushima, 770-8503, Japan.
| | - Yasushi Okuno
- Department of Biomedical Data Intelligence, Kyoto University Graduate School of Medicine, 54 Shogoin Kawahara-Cho, Sakyo-Ku, Kyoto, 606-8507, Japan.
- Department of Artificial Intelligence in Healthcare and Medicine, Kyoto University Graduate School of Medicine, 54 Shogoin Kawahara-Cho, Sakyo-Ku, Kyoto, 606-8507, Japan.
- Biomedical Computational Intelligence Unit, HPC- and AI-Driven Drug Development Platform Division, RIKEN Center for Computational Science, 7-1-26, Minatojima-Minami-Machi, Chuo-Ku, Kobe, Hyogo, 650-0047, Japan.
| |
Collapse
|
24
|
Al-Jipouri A, Eritja À, Bozic M. Unraveling the Multifaceted Roles of Extracellular Vesicles: Insights into Biology, Pharmacology, and Pharmaceutical Applications for Drug Delivery. Int J Mol Sci 2023; 25:485. [PMID: 38203656 PMCID: PMC10779093 DOI: 10.3390/ijms25010485] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 12/19/2023] [Accepted: 12/25/2023] [Indexed: 01/12/2024] Open
Abstract
Extracellular vesicles (EVs) are nanoparticles released from various cell types that have emerged as powerful new therapeutic option for a variety of diseases. EVs are involved in the transmission of biological signals between cells and in the regulation of a variety of biological processes, highlighting them as potential novel targets/platforms for therapeutics intervention and/or delivery. Therefore, it is necessary to investigate new aspects of EVs' biogenesis, biodistribution, metabolism, and excretion as well as safety/compatibility of both unmodified and engineered EVs upon administration in different pharmaceutical dosage forms and delivery systems. In this review, we summarize the current knowledge of essential physiological and pathological roles of EVs in different organs and organ systems. We provide an overview regarding application of EVs as therapeutic targets, therapeutics, and drug delivery platforms. We also explore various approaches implemented over the years to improve the dosage of specific EV products for different administration routes.
Collapse
Affiliation(s)
- Ali Al-Jipouri
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, D-45147 Essen, Germany;
| | - Àuria Eritja
- Vascular and Renal Translational Research Group, Biomedical Research Institute of Lleida Dr. Pifarré Foundation (IRBLLEIDA), 25196 Lleida, Spain;
| | - Milica Bozic
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, D-45147 Essen, Germany;
- Vascular and Renal Translational Research Group, Biomedical Research Institute of Lleida Dr. Pifarré Foundation (IRBLLEIDA), 25196 Lleida, Spain;
| |
Collapse
|
25
|
Lucien F, Gustafson D, Lenassi M, Li B, Teske JJ, Boilard E, von Hohenberg KC, Falcón‐Perez JM, Gualerzi A, Reale A, Jones JC, Lässer C, Lawson C, Nazarenko I, O'Driscoll L, Pink R, Siljander PR, Soekmadji C, Hendrix A, Welsh JA, Witwer KW, Nieuwland R. MIBlood-EV: Minimal information to enhance the quality and reproducibility of blood extracellular vesicle research. J Extracell Vesicles 2023; 12:e12385. [PMID: 38063210 PMCID: PMC10704543 DOI: 10.1002/jev2.12385] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 10/31/2023] [Accepted: 11/10/2023] [Indexed: 12/18/2023] Open
Abstract
Blood is the most commonly used body fluid for extracellular vesicle (EV) research. The composition of a blood sample and its derivatives (i.e., plasma and serum) are not only donor-dependent but also influenced by collection and preparation protocols. Since there are hundreds of pre-analytical protocols and over forty variables, the development of standard operating procedures for EV research is very challenging. To improve the reproducibility of blood EV research, the International Society for Extracellular Vesicles (ISEV) Blood EV Task Force proposes standardized reporting of (i) the applied blood collection and preparation protocol and (ii) the quality of the prepared plasma and serum samples. Gathering detailed information will provide insight into the performance of the protocols and more effectively identify potential confounders in the prepared plasma and serum samples. To collect this information, the ISEV Blood EV Task Force created the Minimal Information for Blood EV research (MIBlood-EV), a tool to record and report information about pre-analytical protocols used for plasma and serum preparation as well as assays used to assess the quality of these preparations. This tool does not require modifications of established local pre-analytical protocols and can be easily implemented to enhance existing databases thereby enabling evidence-based optimization of pre-analytical protocols through meta-analysis. Taken together, insight into the quality of prepared plasma and serum samples will (i) improve the quality of biobanks for EV research, (ii) guide the exchange of plasma and serum samples between biobanks and laboratories, (iii) facilitate inter-laboratory comparative EV studies, and (iv) improve the peer review process.
Collapse
Affiliation(s)
- Fabrice Lucien
- Department of UrologyMayo ClinicRochesterMinnesotaUSA
- Department of ImmunologyMayo ClinicRochesterMinnesotaUSA
| | - Dakota Gustafson
- Department of Laboratory Medicine & PathobiologyUniversity of TorontoTorontoOntarioCanada
- Toronto General Hospital Research InstituteTorontoOntarioCanada
- Department of Public Health SciencesQueen's UniversityKingstonOntarioCanada
| | - Metka Lenassi
- Institute of Biochemistry and Molecular Genetics, Faculty of MedicineUniversity of LjubljanaLjubljanaSlovenia
| | - Bo Li
- Department of Laboratory MedicineNanfang Hospital, Southern Medical UniversityGuangzhouChina
- Laboratory of Experimental Clinical Chemistry, and Amsterdam Vesicle CenterAmsterdam UMClocation AMCAmsterdamNetherlands
| | | | - Eric Boilard
- Centre de Recherche du CHU de Québec – Université Laval, Département de microbiologie et immunologieFaculté de Médecine de l'Université LavalQuébecQuebecCanada
| | | | - Juan Manual Falcón‐Perez
- Exosomes laboratory and Metabolomics PlatformCIC bioGUNE‐BRTADerioSpain
- IKERBASQUE, Basque Foundation for ScienceBilbaoSpain
| | | | - Antonia Reale
- Division of Blood CancersMonash University ‐ Alfred HealthMelbourneVictoriaAustralia
| | - Jennifer C. Jones
- Laboratory of Pathology, Center for Cancer ResearchNational Cancer Institute, National Institutes of HealthBethesdaMarylandUSA
| | - Cecilia Lässer
- Krefting Research Centre, Institute of Medicine at the Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | | | - Irina Nazarenko
- Institute for Infection Prevention and Control, Faculty of MedicineUniversity of Freiburg, Freiburg, Germany, German Cancer Consortium
- Partner Site Freiburg and German Cancer Research CenterHeidelbergGermany
| | - Lorraine O'Driscoll
- School of Pharmacy and Pharmaceutical Sciences & Trinity Biomedical Sciences Institute & Trinity St. James's Cancer InstituteTrinity College DublinDublinIreland
| | - Ryan Pink
- Faculty Health and Life SciencesOxford Brookes UniversityOxfordUnited Kingdom of Great Britain and Northern Ireland
| | - Pia R‐M Siljander
- EV‐group, Molecular and Integrative Biosciences Research Programme, Faculty of Biological and Environmental Sciences, and Drug Research Program, Faculty of Pharmacy, Division of Pharmaceutical BiosciencesUniversity of HelsinkiHelsinkiFinland
| | - Carolina Soekmadji
- School of Biomedical Sciences, Faculty of MedicineUniversity of QueenslandBrisbaneAustralia
| | - An Hendrix
- Laboratory of Experimental Cancer Research, Department of Human Structure and RepairGhent UniversityGhentBelgium
| | - Joshua A Welsh
- School of Medicine, Department of Molecular and Comparative Pathobiology, and Department of NeurologyJohns Hopkins UniversityBaltimoreMarylandUnited States
| | - Kenneth W. Witwer
- School of Medicine, Department of Molecular and Comparative Pathobiology, and Department of NeurologyJohns Hopkins UniversityBaltimoreMarylandUnited States
| | - Rienk Nieuwland
- Laboratory of Experimental Clinical Chemistry, and Amsterdam Vesicle CenterAmsterdam UMClocation AMCAmsterdamNetherlands
| |
Collapse
|
26
|
Spanos M, Gokulnath P, Chatterjee E, Li G, Varrias D, Das S. Expanding the horizon of EV-RNAs: LncRNAs in EVs as biomarkers for disease pathways. EXTRACELLULAR VESICLE 2023; 2:100025. [PMID: 38188000 PMCID: PMC10768935 DOI: 10.1016/j.vesic.2023.100025] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Extracellular vesicles (EVs) are membrane-bound nanoparticles with different types of cargo released by cells and postulated to mediate functions such as intercellular communications. Recent studies have shown that long non-coding RNAs (lncRNAs) or their fragments are present as cargo within EVs. LncRNAs are a heterogeneous group of RNA species with a length exceeding 200 nucleotides with diverse functions in cells based on their localization. While lncRNAs are known for their important functions in cellular regulation, their presence and role in EVs have only recently been explored. While certain studies have observed EV-lncRNAs to be tissue-and disease-specific, it remains to be determined whether or not this is a global observation. Nonetheless, these molecules have demonstrated promising potential to serve as new diagnostic and prognostic biomarkers. In this review, we critically evaluate the role of EV-derived lncRNAs in several prevalent diseases, including cancer, cardiovascular diseases, and neurodegenerative diseases, with a specific focus on their role as biomarkers.
Collapse
Affiliation(s)
- Michail Spanos
- Cardiovascular Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Priyanka Gokulnath
- Cardiovascular Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Emeli Chatterjee
- Cardiovascular Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Guoping Li
- Cardiovascular Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Dimitrios Varrias
- Albert Einstein College of Medicine/Jacobi Medical Center, The Bronx, NY, USA
| | - Saumya Das
- Cardiovascular Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
27
|
Reilly L, Lara E, Ramos D, Li Z, Pantazis CB, Stadler J, Santiana M, Roberts J, Faghri F, Hao Y, Nalls MA, Narayan P, Liu Y, Singleton AB, Cookson MR, Ward ME, Qi YA. A fully automated FAIMS-DIA mass spectrometry-based proteomic pipeline. CELL REPORTS METHODS 2023; 3:100593. [PMID: 37729920 PMCID: PMC10626189 DOI: 10.1016/j.crmeth.2023.100593] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 06/30/2023] [Accepted: 08/24/2023] [Indexed: 09/22/2023]
Abstract
Here, we present a standardized, "off-the-shelf" proteomics pipeline working in a single 96-well plate to achieve deep coverage of cellular proteomes with high throughput and scalability. This integrated pipeline streamlines a fully automated sample preparation platform, a data-independent acquisition (DIA) coupled with high-field asymmetric waveform ion mobility spectrometer (FAIMS) interface, and an optimized library-free DIA database search strategy. Our systematic evaluation of FAIMS-DIA showing single compensation voltage (CV) at -35 V not only yields the deepest proteome coverage but also best correlates with DIA without FAIMS. Our in-depth comparison of direct-DIA database search engines shows that Spectronaut outperforms others, providing the highest quantifiable proteins. Next, we apply three common DIA strategies in characterizing human induced pluripotent stem cell (iPSC)-derived neurons and show single-shot mass spectrometry (MS) using single-CV (-35 V)-FAIMS-DIA results in >9,000 quantifiable proteins with <10% missing values, as well as superior reproducibility and accuracy compared with other existing DIA methods.
Collapse
Affiliation(s)
- Luke Reilly
- Center for Alzheimer's and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Erika Lara
- Center for Alzheimer's and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Daniel Ramos
- Center for Alzheimer's and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Ziyi Li
- Center for Alzheimer's and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA; Data Tecnica International, LLC, Glen Echo, MD, USA
| | - Caroline B Pantazis
- Center for Alzheimer's and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Julia Stadler
- Center for Alzheimer's and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Marianita Santiana
- Center for Alzheimer's and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Jessica Roberts
- Center for Alzheimer's and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Faraz Faghri
- Center for Alzheimer's and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA; Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA; Data Tecnica International, LLC, Glen Echo, MD, USA
| | - Ying Hao
- Center for Alzheimer's and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Mike A Nalls
- Center for Alzheimer's and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA; Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA; Data Tecnica International, LLC, Glen Echo, MD, USA
| | - Priyanka Narayan
- Center for Alzheimer's and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA; Genetics and Biochemistry Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20814, USA
| | - Yansheng Liu
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Andrew B Singleton
- Center for Alzheimer's and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA; Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Mark R Cookson
- Center for Alzheimer's and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA; Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Michael E Ward
- Center for Alzheimer's and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA; National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Yue A Qi
- Center for Alzheimer's and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
28
|
Xylaki M, Chopra A, Weber S, Bartl M, Outeiro TF, Mollenhauer B. Extracellular Vesicles for the Diagnosis of Parkinson's Disease: Systematic Review and Meta-Analysis. Mov Disord 2023; 38:1585-1597. [PMID: 37449706 DOI: 10.1002/mds.29497] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 05/23/2023] [Accepted: 05/24/2023] [Indexed: 07/18/2023] Open
Abstract
Parkinson's disease (PD) biomarkers are needed by both clinicians and researchers (for diagnosis, identifying study populations, and monitoring therapeutic response). Imaging, genetic, and biochemical biomarkers have been widely studied. In recent years, extracellular vesicles (EVs) have become a promising material for biomarker development. Proteins and molecular material from any organ, including the central nervous system, can be packed into EVs and transported to the periphery into easily obtainable biological specimens like blood, urine, and saliva. We performed a systematic review and meta-analysis of articles (published before November 15, 2022) reporting biomarker assessment in EVs in PD patients and healthy controls (HCs). Biomarkers were analyzed using random effects meta-analysis and the calculated standardized mean difference (Std.MD). Several proteins and ribonucleic acids have been identified in EVs in PD patients, but only α-synuclein (aSyn) and leucine-rich repeat kinase 2 (LRRK2) were reported in sufficient studies (n = 24 and 6, respectively) to perform a meta-analysis. EV aSyn was significantly increased in neuronal L1 cell adhesion molecule (L1CAM)-positive blood EVs in PD patients compared to HCs (Std.MD = 1.84, 95% confidence interval = 0.76-2.93, P = 0.0009). Further analysis of the biological sample and EV isolation method indicated that L1CAM-IP (immunoprecipitation) directly from plasma was the best isolation method for assessing aSyn in PD patients. Upcoming neuroprotective clinical trials immediately need peripheral biomarkers for identifying individuals at risk of developing PD. Overall, the improved sensitivity of assays means they can identify biomarkers in blood that reflect changes in the brain. CNS-derived EVs in blood will likely play a major role in biomarker development in the coming years. © 2023 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Mary Xylaki
- Department of Neurology, University Medical Center Goettingen, Goettingen, Germany
| | - Avika Chopra
- Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Goettingen, Goettingen, Germany
| | - Sandrina Weber
- Department of Neurology, University Medical Center Goettingen, Goettingen, Germany
| | - Michael Bartl
- Department of Neurology, University Medical Center Goettingen, Goettingen, Germany
| | - Tiago F Outeiro
- Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Goettingen, Goettingen, Germany
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle, Upon Tyne, United Kingdom
- Max Planck Institute for Multidisciplinary Sciences, Goettingen, Germany
- Scientific Employee with an Honorary Contract at German Center for Neurodegenerative Diseases (DZNE), Goettingen, Germany
| | - Brit Mollenhauer
- Department of Neurology, University Medical Center Goettingen, Goettingen, Germany
- Scientific Employee with an Honorary Contract at German Center for Neurodegenerative Diseases (DZNE), Goettingen, Germany
- Paracelsus-Elena-Klinik, Kassel, Germany
| |
Collapse
|
29
|
Hu L, Liu X, Zheng Q, Chen W, Xu H, Li H, Luo J, Yang R, Mao X, Wang S, Chen T, Lee LP, Liu F. Interaction network of extracellular vesicles building universal analysis via eye tears: iNEBULA. SCIENCE ADVANCES 2023; 9:eadg1137. [PMID: 36921051 PMCID: PMC10017052 DOI: 10.1126/sciadv.adg1137] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 02/15/2023] [Indexed: 06/18/2023]
Abstract
Discovering the secrets of diseases from tear extracellular vesicles (EVs) is well-recognized and appreciated. However, a precise understanding of the interaction network between EV populations and their biogenesis from our body requires more in-depth and systematic analysis. Here, we report the biological profiles of different-size tear EV subsets from healthy individuals and the origins of EV proteins. We have identified about 1800 proteins and revealed the preferential differences in the biogenesis among distinct subsets. We observe that eye-related proteins that maintain retinal homeostasis and regulate inflammation are preferentially enriched in medium-size EVs (100 to 200 nm) fractions. Using universal analysis in combination with the Human Protein Atlas consensus dataset, we found the genesis of tear EV proteins with 37 tissues and 79 cell types. The proteins related to retinal neuronal cells, glial cells, and blood and immune cells are selectively enriched among EV subsets. Our studies in heterogeneous tear EVs provide building blocks for future transformative precision molecular diagnostics and therapeutics.
Collapse
Affiliation(s)
- Liang Hu
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Xiaoling Liu
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Qiaolan Zheng
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Wuhe Chen
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Hao Xu
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Hengrui Li
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Jiaxin Luo
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Rui Yang
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Xulong Mao
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Siyao Wang
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Tucan Chen
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Luke P. Lee
- Renal Division and Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Department of Bioengineering, University of California at Berkeley, Berkeley, CA 94720, USA
- Department of Electrical Engineering and Computer Science, University of California at Berkeley, Berkeley, CA 94720, USA
- Institute of Quantum Biophysics, Department of Biophysics, Sungkyunkwan University, Suwon, Gyeonggi-do 16419, Korea
| | - Fei Liu
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
| |
Collapse
|
30
|
Xu G, Lv J, Huang M, Zhu L, Tan S, Ding C. Comparison of Pairwise Venous and Fingertip Plasma Using Quantitative Proteomics Based on Data-Independent Acquisition. J Proteome Res 2023; 22:1347-1358. [PMID: 36882937 DOI: 10.1021/acs.jproteome.3c00081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/09/2023]
Abstract
Blood contains a great deal of health-related information and can be used to monitor human health status. Clinically, venous or fingertip blood is usually used for blood tests. However, the clinical application settings of the two sources of blood are unclear. In this study, the proteomes of pairwise venous plasma (VP) and fingertip plasma (FP) were analyzed, and the levels of 3797 proteins were compared between VP and FP. The Spearman's correlation coefficient for the relationship between protein levels of VP and FP ranges from 0.64 to 0.78 (p < 0.0001). The common pathways of VP and FP are related to cell-cell adhesion, protein stabilization, innate immune response, and complement activation, the classical pathway. The VP-overrepresented pathway is related to actin filament organization, while the FP-overrepresented pathway is related to the hydrogen peroxide catabolic process. ADAMTSL4, ADIPOQ, HIBADH, and XPO5 both in VP and FP are potential gender-related proteins. Notably, the VP proteome has a higher interpretation on age than the FP proteome, and CD14 is a potential age-related protein in VP but not in FP. Our study mapped the different proteomes between VP and FP, which can provide value for the standardization of clinical blood tests.
Collapse
Affiliation(s)
- Ganfei Xu
- State Key Laboratory of Genetic Engineering and Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Institute of Biomedical Sciences, Human Phenome Institute, Zhongshan Hospital, Fudan University, Shanghai 200433, China
| | - Jiacheng Lv
- State Key Laboratory of Genetic Engineering and Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Institute of Biomedical Sciences, Human Phenome Institute, Zhongshan Hospital, Fudan University, Shanghai 200433, China
| | - Mingjing Huang
- State Key Laboratory of Genetic Engineering and Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Institute of Biomedical Sciences, Human Phenome Institute, Zhongshan Hospital, Fudan University, Shanghai 200433, China
| | - Lingli Zhu
- State Key Laboratory of Genetic Engineering and Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Institute of Biomedical Sciences, Human Phenome Institute, Zhongshan Hospital, Fudan University, Shanghai 200433, China
| | - Subei Tan
- State Key Laboratory of Genetic Engineering and Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Institute of Biomedical Sciences, Human Phenome Institute, Zhongshan Hospital, Fudan University, Shanghai 200433, China
| | - Chen Ding
- State Key Laboratory of Genetic Engineering and Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Institute of Biomedical Sciences, Human Phenome Institute, Zhongshan Hospital, Fudan University, Shanghai 200433, China.,State Key Laboratory of Cell Differentiation and Regulation, Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Center for Outstanding Overseas Scientists of Pulmonary Fibrosis, College of Life Science, Institute of Biomedical Science, Henan Normal University, Xinxiang 453007, China.,Academy of Medical Science, Zhengzhou University, Zhengzhou 450052, China
| |
Collapse
|
31
|
Liu Y, Li Y, Zeng T. Multi-omics of extracellular vesicles: An integrative representation of functional mediators and perspectives on lung disease study. FRONTIERS IN BIOINFORMATICS 2023; 3:1117271. [PMID: 36844931 PMCID: PMC9947558 DOI: 10.3389/fbinf.2023.1117271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 01/31/2023] [Indexed: 02/11/2023] Open
Abstract
Extracellular vesicles are secreted by almost all cell types. EVs include a broader component known as exosomes that participate in cell-cell and tissue-tissue communication via carrying diverse biological signals from one cell type or tissue to another. EVs play roles as communication messengers of the intercellular network to mediate different physiological activities or pathological changes. In particular, most EVs are natural carriers of functional cargo such as DNA, RNA, and proteins, and thus they are relevant to advancing personalized targeted therapies in clinical practice. For the application of EVs, novel bioinformatic models and methods based on high-throughput technologies and multi-omics data are required to provide a deeper understanding of their biological and biomedical characteristics. These include qualitative and quantitative representation for identifying cargo markers, local cellular communication inference for tracing the origin and production of EVs, and distant organ communication reconstruction for targeting the influential microenvironment and transferable activators. Thus, this perspective paper introduces EVs in the context of multi-omics and provides an integrative bioinformatic viewpoint of the state of current research on EVs and their applications.
Collapse
Affiliation(s)
| | - Yixue Li
- *Correspondence: Yixue Li, ; Tao Zeng,
| | - Tao Zeng
- *Correspondence: Yixue Li, ; Tao Zeng,
| |
Collapse
|
32
|
Kawasaki T, Takeda Y, Edahiro R, Shirai Y, Nogami-Itoh M, Matsuki T, Kida H, Enomoto T, Hara R, Noda Y, Adachi Y, Niitsu T, Amiya S, Yamaguchi Y, Murakami T, Kato Y, Morita T, Yoshimura H, Yamamoto M, Nakatsubo D, Miyake K, Shiroyama T, Hirata H, Adachi J, Okada Y, Kumanogoh A. Next-generation proteomics of serum extracellular vesicles combined with single-cell RNA sequencing identifies MACROH2A1 associated with refractory COVID-19. Inflamm Regen 2022; 42:53. [PMID: 36451245 PMCID: PMC9709739 DOI: 10.1186/s41232-022-00243-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 11/18/2022] [Indexed: 12/02/2022] Open
Abstract
BACKGROUND The coronavirus disease 2019 (COVID-19) pandemic is widespread; however, accurate predictors of refractory cases have not yet been established. Circulating extracellular vesicles, involved in many pathological processes, are ideal resources for biomarker exploration. METHODS To identify potential serum biomarkers and examine the proteins associated with the pathogenesis of refractory COVID-19, we conducted high-coverage proteomics on serum extracellular vesicles collected from 12 patients with COVID-19 at different disease severity levels and 4 healthy controls. Furthermore, single-cell RNA sequencing of peripheral blood mononuclear cells collected from 10 patients with COVID-19 and 5 healthy controls was performed. RESULTS Among the 3046 extracellular vesicle proteins that were identified, expression of MACROH2A1 was significantly elevated in refractory cases compared to non-refractory cases; moreover, its expression was increased according to disease severity. In single-cell RNA sequencing of peripheral blood mononuclear cells, the expression of MACROH2A1 was localized to monocytes and elevated in critical cases. Consistently, single-nucleus RNA sequencing of lung tissues revealed that MACROH2A1 was highly expressed in monocytes and macrophages and was significantly elevated in fatal COVID-19. Moreover, molecular network analysis showed that pathways such as "estrogen signaling pathway," "p160 steroid receptor coactivator (SRC) signaling pathway," and "transcriptional regulation by STAT" were enriched in the transcriptome of monocytes in the peripheral blood mononuclear cells and lungs, and they were also commonly enriched in extracellular vesicle proteomics. CONCLUSIONS Our findings highlight that MACROH2A1 in extracellular vesicles is a potential biomarker of refractory COVID-19 and may reflect the pathogenesis of COVID-19 in monocytes.
Collapse
Affiliation(s)
- Takahiro Kawasaki
- grid.136593.b0000 0004 0373 3971Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Osaka, 565-0871 Japan ,grid.136593.b0000 0004 0373 3971Laboratory of Immunopathology, World Premier International Immunology Frontier Research Center (WPI-IFReC), Osaka University, Osaka, 565-0871 Japan
| | - Yoshito Takeda
- grid.136593.b0000 0004 0373 3971Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Osaka, 565-0871 Japan
| | - Ryuya Edahiro
- grid.136593.b0000 0004 0373 3971Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Osaka, 565-0871 Japan ,grid.136593.b0000 0004 0373 3971Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, Japan
| | - Yuya Shirai
- grid.136593.b0000 0004 0373 3971Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Osaka, 565-0871 Japan ,grid.136593.b0000 0004 0373 3971Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, Japan
| | - Mari Nogami-Itoh
- grid.482562.fLaboratory of Bioinformatics, Artificial Intelligence Center for Health and Biomedical Research, National Institutes of Biomedical Innovation, Health and Nutrition, 7-6-8 Saito-Asagi, Ibaraki, Osaka 567-0085 Japan
| | - Takanori Matsuki
- grid.416803.80000 0004 0377 7966Department of Respiratory Medicine, National Hospital Organization Osaka Toneyama Medical Center, 5-1-1 Toneyama, Toyonaka, Osaka 560-8552 Japan
| | - Hiroshi Kida
- grid.416803.80000 0004 0377 7966Department of Respiratory Medicine, National Hospital Organization Osaka Toneyama Medical Center, 5-1-1 Toneyama, Toyonaka, Osaka 560-8552 Japan
| | - Takatoshi Enomoto
- grid.136593.b0000 0004 0373 3971Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Osaka, 565-0871 Japan
| | - Reina Hara
- grid.136593.b0000 0004 0373 3971Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Osaka, 565-0871 Japan
| | - Yoshimi Noda
- grid.136593.b0000 0004 0373 3971Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Osaka, 565-0871 Japan
| | - Yuichi Adachi
- grid.136593.b0000 0004 0373 3971Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Osaka, 565-0871 Japan
| | - Takayuki Niitsu
- grid.136593.b0000 0004 0373 3971Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Osaka, 565-0871 Japan
| | - Saori Amiya
- grid.136593.b0000 0004 0373 3971Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Osaka, 565-0871 Japan
| | - Yuta Yamaguchi
- grid.136593.b0000 0004 0373 3971Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Osaka, 565-0871 Japan
| | - Teruaki Murakami
- grid.136593.b0000 0004 0373 3971Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Osaka, 565-0871 Japan
| | - Yasuhiro Kato
- grid.136593.b0000 0004 0373 3971Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Osaka, 565-0871 Japan
| | - Takayoshi Morita
- grid.136593.b0000 0004 0373 3971Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Osaka, 565-0871 Japan
| | - Hanako Yoshimura
- grid.136593.b0000 0004 0373 3971Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Osaka, 565-0871 Japan
| | - Makoto Yamamoto
- grid.136593.b0000 0004 0373 3971Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Osaka, 565-0871 Japan
| | - Daisuke Nakatsubo
- grid.136593.b0000 0004 0373 3971Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Osaka, 565-0871 Japan
| | - Kotaro Miyake
- grid.136593.b0000 0004 0373 3971Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Osaka, 565-0871 Japan
| | - Takayuki Shiroyama
- grid.136593.b0000 0004 0373 3971Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Osaka, 565-0871 Japan
| | - Haruhiko Hirata
- grid.136593.b0000 0004 0373 3971Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Osaka, 565-0871 Japan
| | - Jun Adachi
- grid.482562.fLaboratory of Proteomics for Drug Discovery, Center for Drug Design Research, National Institute of Biomedical Innovation, Health and Nutrition, 7-6-8, Saito-Asagi, Ibaraki City, Osaka, 567-0085 Japan
| | - Yukinori Okada
- grid.136593.b0000 0004 0373 3971Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, Japan
| | - Atsushi Kumanogoh
- grid.136593.b0000 0004 0373 3971Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Osaka, 565-0871 Japan ,grid.136593.b0000 0004 0373 3971Laboratory of Immunopathology, World Premier International Immunology Frontier Research Center (WPI-IFReC), Osaka University, Osaka, 565-0871 Japan ,grid.136593.b0000 0004 0373 3971Center for Infectious Diseases for Education and Research (CiDER), Osaka University, Suita, Osaka Japan ,grid.136593.b0000 0004 0373 3971Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives (OTRI), Osaka University, Suita, Japan ,grid.480536.c0000 0004 5373 4593Japan Agency for Medical Research and Development-Core Research for Evolutionary Medical Science and Technology (AMED-CREST), Japan Agency for Medical Research and Development, Tokyo, Japan ,grid.136593.b0000 0004 0373 3971Center for Advanced Modalities and DDS (CAMaD), Osaka University, Osaka, Japan
| |
Collapse
|
33
|
Muraoka S, Hirano M, Isoyama J, Ishida M, Tomonaga T, Adachi J. Automated Proteomics Sample Preparation of Phosphatidylserine-Positive Extracellular Vesicles from Human Body Fluids. ACS OMEGA 2022; 7:41472-41479. [PMID: 36406491 PMCID: PMC9670285 DOI: 10.1021/acsomega.2c05244] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 10/24/2022] [Indexed: 06/16/2023]
Abstract
Extracellular vesicles (EVs) are ubiquitously secreted by almost every cell type and are present in all body fluids. Blood-derived EVs can be used as a promising source for biomarker monitoring in disease. EV proteomics is currently being analyzed in clinical specimens. However, their EV proteomics preparation methods are limited in throughput for human subjects. Here, we introduced a novel automated EV isolation and sample preparation method using a magnetic particle processing robot for automated 96-well processing of magnetic particles for EV proteomics analysis that can be started with a low volume of multiple clinical samples. The automation of EV purification reduced the coefficient of variation of protein quantification from 3.5 to 2.2% compared with manual purification, enabling the quantification of 1120 proteins in 1 h of MS analysis. This automated proteomics EV sample preparation is attractive for processing large cohort samples for biomarker development, validation, and routine testing.
Collapse
Affiliation(s)
- Satoshi Muraoka
- Laboratory
of Proteomics for Drug Discovery, Center for Drug Design Research, National Institute of Biomedical Innovation, Health
and Nutrition, Osaka 567-0085, Japan
- Laboratory
of Clinical and Analytical Chemistry, Collaborative Research Center
for Health and Medicine, National Institute
of Biomedical Innovation, Health and Nutrition, Osaka 567-0085, Japan
| | - Masayo Hirano
- Laboratory
of Proteomics for Drug Discovery, Center for Drug Design Research, National Institute of Biomedical Innovation, Health
and Nutrition, Osaka 567-0085, Japan
| | - Junko Isoyama
- Laboratory
of Proteomics for Drug Discovery, Center for Drug Design Research, National Institute of Biomedical Innovation, Health
and Nutrition, Osaka 567-0085, Japan
| | - Mimiko Ishida
- Laboratory
of Proteomics for Drug Discovery, Center for Drug Design Research, National Institute of Biomedical Innovation, Health
and Nutrition, Osaka 567-0085, Japan
| | - Takeshi Tomonaga
- Laboratory
of Proteomics for Drug Discovery, Center for Drug Design Research, National Institute of Biomedical Innovation, Health
and Nutrition, Osaka 567-0085, Japan
| | - Jun Adachi
- Laboratory
of Proteomics for Drug Discovery, Center for Drug Design Research, National Institute of Biomedical Innovation, Health
and Nutrition, Osaka 567-0085, Japan
- Laboratory
of Clinical and Analytical Chemistry, Collaborative Research Center
for Health and Medicine, National Institute
of Biomedical Innovation, Health and Nutrition, Osaka 567-0085, Japan
- Laboratory
of Proteomics and Drug Discovery, Graduate School of Pharmaceutical
Sciences, Kyoto University, Kyoto 606-8501, Japan
| |
Collapse
|
34
|
Extracellular vesicle isolation, purification and evaluation in cancer diagnosis. Expert Rev Mol Med 2022; 24:e41. [PMID: 36268744 DOI: 10.1017/erm.2022.34] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Strategies for non-invasive biomarker discovery in early detection of cancer are an urgent need. Extracellular vesicles (EVs) have generated increasing attention from the scientific community and are under intensive investigations due to their unique biological profiles and their non-invasive nature. EVs are membrane-enclosed vesicles with variable sizes and function. Such vesicles are actively secreted from multiple cell types and are considered as key vehicles for inter-cellular communications and signalling. The stability and potential to easily cross biological barriers enable EVs for exerting durable effects on target cells. These along with easy access to such vesicles, the consistent secretion from tumour during all stages of tumorigenesis and their content providing a reservoir of molecules as well as mirroring the identity of the cell of origin are virtues that have made EVs appealing to be assessed in liquid biopsy approaches and for using as a promising resource of biomarkers in cancer diagnosis and therapy and monitoring targeted cancer therapy. Early detection of EVs will guide time-scheduled personalised therapy. Surveying reliable and sensitive methods for rapid isolation of EVs from biofluids, the purity of isolated vesicles and their molecular profiling and marker specification for clinical translation in patients with cancer are issues in the area and the hot topics of many recent studies. Here, the focus is over methods for EV isolation and stratification for digging more information about liquid biopsy-based diagnosis. Extending knowledge regarding EV-based strategies is a key to validate independent patient follow-up for cancer diagnosis at early stages and inspecting the efficacy of therapeutics.
Collapse
|
35
|
Hullin-Matsuda F, Colosetti P, Rabia M, Luquain-Costaz C, Delton I. Exosomal lipids from membrane organization to biomarkers: Focus on an endolysosomal-specific lipid. Biochimie 2022; 203:77-92. [DOI: 10.1016/j.biochi.2022.09.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 09/22/2022] [Accepted: 09/26/2022] [Indexed: 11/30/2022]
|
36
|
Shaba E, Vantaggiato L, Governini L, Haxhiu A, Sebastiani G, Fignani D, Grieco GE, Bergantini L, Bini L, Landi C. Multi-Omics Integrative Approach of Extracellular Vesicles: A Future Challenging Milestone. Proteomes 2022; 10:proteomes10020012. [PMID: 35645370 PMCID: PMC9149947 DOI: 10.3390/proteomes10020012] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 04/14/2022] [Accepted: 04/19/2022] [Indexed: 02/01/2023] Open
Abstract
In the era of multi-omic sciences, dogma on singular cause-effect in physio-pathological processes is overcome and system biology approaches have been providing new perspectives to see through. In this context, extracellular vesicles (EVs) are offering a new level of complexity, given their role in cellular communication and their activity as mediators of specific signals to target cells or tissues. Indeed, their heterogeneity in terms of content, function, origin and potentiality contribute to the cross-interaction of almost every molecular process occurring in a complex system. Such features make EVs proper biological systems being, therefore, optimal targets of omic sciences. Currently, most studies focus on dissecting EVs content in order to either characterize it or to explore its role in various pathogenic processes at transcriptomic, proteomic, metabolomic, lipidomic and genomic levels. Despite valuable results being provided by individual omic studies, the categorization of EVs biological data might represent a limit to be overcome. For this reason, a multi-omic integrative approach might contribute to explore EVs function, their tissue-specific origin and their potentiality. This review summarizes the state-of-the-art of EVs omic studies, addressing recent research on the integration of EVs multi-level biological data and challenging developments in EVs origin.
Collapse
Affiliation(s)
- Enxhi Shaba
- Functional Proteomics Lab, Department of Life Sciences, University of Siena, 53100 Siena, Italy; (L.V.); (L.B.); (C.L.)
- Correspondence:
| | - Lorenza Vantaggiato
- Functional Proteomics Lab, Department of Life Sciences, University of Siena, 53100 Siena, Italy; (L.V.); (L.B.); (C.L.)
| | - Laura Governini
- Department of Molecular and Developmental Medicine, University of Siena, 53100 Siena, Italy; (L.G.); (A.H.)
| | - Alesandro Haxhiu
- Department of Molecular and Developmental Medicine, University of Siena, 53100 Siena, Italy; (L.G.); (A.H.)
| | - Guido Sebastiani
- Diabetes Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, 53100 Siena, Italy; (G.S.); (D.F.); (G.E.G.)
- Fondazione Umberto Di Mario, c/o Toscana Life Sciences, 53100 Siena, Italy
| | - Daniela Fignani
- Diabetes Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, 53100 Siena, Italy; (G.S.); (D.F.); (G.E.G.)
- Fondazione Umberto Di Mario, c/o Toscana Life Sciences, 53100 Siena, Italy
| | - Giuseppina Emanuela Grieco
- Diabetes Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, 53100 Siena, Italy; (G.S.); (D.F.); (G.E.G.)
- Fondazione Umberto Di Mario, c/o Toscana Life Sciences, 53100 Siena, Italy
| | - Laura Bergantini
- Respiratory Diseases and Lung Transplant Unit, Department of Medical Sciences, Surgery and Neurosciences, University of Siena, 53100 Siena, Italy;
| | - Luca Bini
- Functional Proteomics Lab, Department of Life Sciences, University of Siena, 53100 Siena, Italy; (L.V.); (L.B.); (C.L.)
| | - Claudia Landi
- Functional Proteomics Lab, Department of Life Sciences, University of Siena, 53100 Siena, Italy; (L.V.); (L.B.); (C.L.)
| |
Collapse
|