1
|
Li Y, Wang Y, Zhang Y, Zhu Y, Dong Y, Cheng H, Zhang Y, Wang Y, Li Z, Gao J. Engineered mesenchymal stem cell-derived extracellular vesicles: kill tumors and protect organs. Theranostics 2024; 14:6202-6217. [PMID: 39431009 PMCID: PMC11488101 DOI: 10.7150/thno.99618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 08/20/2024] [Indexed: 10/22/2024] Open
Abstract
Solid tumors cause 90% of cancers and remain the primary cause of mortality. However, treating solid tumors presents significant challenges due to the complex tumor microenvironment and drug resistance, leading to inadequate treatment targeting and severe side effects. Surgery, radiotherapy, and chemotherapy Although it is an effective method for the treatment of solid tumors, it can lead to organ dysfunction and affect patient prognosis. Therefore, it is imperative to improve treatment precision and organ repair capabilities to manage solid tumors. Mesenchymal stem cell extracellular vesicles (MSC-EVs) have wide application prospects as a new agent for solid tumor therapy. Firstly, MSC-EVs is a derivative of MSCs. It has the function of promoting tissue regeneration by inducing dedifferentiation in surviving cells after injury. Additionally, MSC-EVs offer unique advantages in terms of safety, stability and penetrability, making them a promising extracellular therapeutic modality for solid tumor treatment. Finally, MSC-EVs are able to enhance therapeutic efficacy through engineering strategies. To sum up, this review takes MSC-EVs as its object. And then we discuss recent advancements and engineering strategies in the use of MSC-EVs for soid tumor suppression. This review aims to inspire researchers to devise a new method for effectively treat solid tumors.
Collapse
Affiliation(s)
- Yu Li
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
- Stem Cell and Regeneration Medicine Institute, Research Center of Translational Medicine, Naval Medical University, Shanghai, 200433, China
- Changhai Clinical Research Unit, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Yao Wang
- Changhai Clinical Research Unit, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
- College of Life Science, Mudanjiang Medical University, Heilongjiang Mudanjiang, 157011, China
| | - Yu Zhang
- Shanghai Key Laboratory of Cell Engineering, Shanghai, 200120, China
| | - Yuruchen Zhu
- School of Basic Medical Sciences, Naval Medical University, Shanghai, 200433, China
| | - Yuhui Dong
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, 200093, China
| | - Haobin Cheng
- School of Basic Medical Sciences, Naval Medical University, Shanghai, 200433, China
| | - Yinan Zhang
- School of Chemical Science and Engineering, Tongji University, Shanghai, 200092, China
| | - Yue Wang
- Stem Cell and Regeneration Medicine Institute, Research Center of Translational Medicine, Naval Medical University, Shanghai, 200433, China
- Shanghai Key Laboratory of Cell Engineering, Shanghai, 200120, China
- Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai, 200120, China
| | - Zhaoshen Li
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
- Changhai Clinical Research Unit, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
- National Clinical Research Center for Digestive Diseases, Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
- National Key Laboratory of lmmunology and Inflammation, Naval Medical University, Shanghai, 200433, China
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
- Shanghai Key Laboratory of Nautical Medicine and Translation of Drugs and Medical Devices, Shanghai, 200433, China
| | - Jie Gao
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
- Changhai Clinical Research Unit, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
- Shanghai Key Laboratory of Nautical Medicine and Translation of Drugs and Medical Devices, Shanghai, 200433, China
| |
Collapse
|
2
|
Zhao W, Li K, Li L, Wang R, Lei Y, Yang H, Sun L. Mesenchymal Stem Cell-Derived Exosomes as Drug Delivery Vehicles in Disease Therapy. Int J Mol Sci 2024; 25:7715. [PMID: 39062956 PMCID: PMC11277139 DOI: 10.3390/ijms25147715] [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: 06/07/2024] [Revised: 07/09/2024] [Accepted: 07/11/2024] [Indexed: 07/28/2024] Open
Abstract
Exosomes are small vesicles containing proteins, nucleic acids, and biological lipids, which are responsible for intercellular communication. Studies have shown that exosomes can be utilized as effective drug delivery vehicles to accurately deliver therapeutic substances to target tissues, enhancing therapeutic effects and reducing side effects. Mesenchymal stem cells (MSCs) are a class of stem cells widely used for tissue engineering, regenerative medicine, and immunotherapy. Exosomes derived from MSCs have special immunomodulatory functions, low immunogenicity, the ability to penetrate tumor tissues, and high yield, which are expected to be engineered into efficient drug delivery systems. Despite the promising promise of MSC-derived exosomes, exploring their optimal preparation methods, drug-loading modalities, and therapeutic potential remains challenging. Therefore, this article reviews the related characteristics, preparation methods, application, and potential risks of MSC-derived exosomes as drug delivery systems in order to find potential therapeutic breakthroughs.
Collapse
Affiliation(s)
- Wenzhe Zhao
- School of Life Sciences, Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment in Special Environment, Northwestern Polytechnical University, Xi’an 710072, China; (W.Z.); (K.L.); (L.L.); (R.W.); (Y.L.)
- Dongguan Sanhang Innovation Institute, Dongguan 523808, China
| | - Kaixuan Li
- School of Life Sciences, Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment in Special Environment, Northwestern Polytechnical University, Xi’an 710072, China; (W.Z.); (K.L.); (L.L.); (R.W.); (Y.L.)
- Dongguan Sanhang Innovation Institute, Dongguan 523808, China
| | - Liangbo Li
- School of Life Sciences, Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment in Special Environment, Northwestern Polytechnical University, Xi’an 710072, China; (W.Z.); (K.L.); (L.L.); (R.W.); (Y.L.)
- Dongguan Sanhang Innovation Institute, Dongguan 523808, China
| | - Ruichen Wang
- School of Life Sciences, Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment in Special Environment, Northwestern Polytechnical University, Xi’an 710072, China; (W.Z.); (K.L.); (L.L.); (R.W.); (Y.L.)
- Dongguan Sanhang Innovation Institute, Dongguan 523808, China
| | - Yang Lei
- School of Life Sciences, Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment in Special Environment, Northwestern Polytechnical University, Xi’an 710072, China; (W.Z.); (K.L.); (L.L.); (R.W.); (Y.L.)
- Dongguan Sanhang Innovation Institute, Dongguan 523808, China
| | - Hui Yang
- School of Life Sciences, Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment in Special Environment, Northwestern Polytechnical University, Xi’an 710072, China; (W.Z.); (K.L.); (L.L.); (R.W.); (Y.L.)
| | - Leming Sun
- School of Life Sciences, Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment in Special Environment, Northwestern Polytechnical University, Xi’an 710072, China; (W.Z.); (K.L.); (L.L.); (R.W.); (Y.L.)
- Dongguan Sanhang Innovation Institute, Dongguan 523808, China
| |
Collapse
|
3
|
Martínez-Díaz P, Parra A, Sanchez-López CM, Casas J, Lucas X, Marcilla A, Roca J, Barranco I. Small and Large Extracellular Vesicles of Porcine Seminal Plasma Differ in Lipid Profile. Int J Mol Sci 2024; 25:7492. [PMID: 39000599 PMCID: PMC11242203 DOI: 10.3390/ijms25137492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 07/03/2024] [Accepted: 07/05/2024] [Indexed: 07/16/2024] Open
Abstract
Seminal plasma contains a heterogeneous population of extracellular vesicles (sEVs) that remains poorly characterized. This study aimed to characterize the lipidomic profile of two subsets of differently sized sEVs, small (S-) and large (L-), isolated from porcine seminal plasma by size-exclusion chromatography and characterized by an orthogonal approach. High-performance liquid chromatography-high-resolution mass spectrometry was used for lipidomic analysis. A total of 157 lipid species from 14 lipid classes of 4 major categories (sphingolipids, glycerophospholipids, glycerolipids, and sterols) were identified. Qualitative differences were limited to two cholesteryl ester species present only in S-sEVs. L-sEVs had higher levels of all quantified lipid classes due to their larger membrane surface area. The distribution pattern was different, especially for sphingomyelins (more in S-sEVs) and ceramides (more in L-sEVs). In conclusion, this study reveals differences in the lipidomic profile of two subsets of porcine sEVs, suggesting that they differ in biogenesis and functionality.
Collapse
Affiliation(s)
- Pablo Martínez-Díaz
- Department of Medicine and Animal Surgery, Faculty of Veterinary Science, University of Murcia, 30100 Murcia, Spain
| | - Ana Parra
- Department of Medicine and Animal Surgery, Faculty of Veterinary Science, University of Murcia, 30100 Murcia, Spain
| | - Christian M Sanchez-López
- Àrea de Parasitologia, Departament de Farmàcia i Tecnologia Farmacèutica i Parasitologia, Universitat de València, 46100 Valencia, Spain
- Joint Research Unit on Endocrinology, Nutrition and Clinical Dietetics, Health Research Institute La Fe, Universitat de València, 46100 Valencia, Spain
| | - Josefina Casas
- Research Unit on BioActive Molecules (RUBAM), Institute for Advanced Chemistry (IQAC-CSIC), Jordi Girona 18-26, 08034 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Xiomara Lucas
- Department of Medicine and Animal Surgery, Faculty of Veterinary Science, University of Murcia, 30100 Murcia, Spain
| | - Antonio Marcilla
- Àrea de Parasitologia, Departament de Farmàcia i Tecnologia Farmacèutica i Parasitologia, Universitat de València, 46100 Valencia, Spain
- Joint Research Unit on Endocrinology, Nutrition and Clinical Dietetics, Health Research Institute La Fe, Universitat de València, 46100 Valencia, Spain
| | - Jordi Roca
- Department of Medicine and Animal Surgery, Faculty of Veterinary Science, University of Murcia, 30100 Murcia, Spain
| | - Isabel Barranco
- Department of Medicine and Animal Surgery, Faculty of Veterinary Science, University of Murcia, 30100 Murcia, Spain
| |
Collapse
|
4
|
Zhang B, Lai RC, Sim WK, Tan TT, Lim SK. An Assessment of Administration Route on MSC-sEV Therapeutic Efficacy. Biomolecules 2024; 14:622. [PMID: 38927026 PMCID: PMC11202284 DOI: 10.3390/biom14060622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 05/15/2024] [Accepted: 05/22/2024] [Indexed: 06/28/2024] Open
Abstract
Mesenchymal stem/stromal cell-derived small extracellular vesicles (MSC-sEVs) are promising therapeutic agents. In this study, we investigated how the administration route of MSC-sEVs affects their therapeutic efficacy in a mouse model of bleomycin (BLM)-induced skin scleroderma (SSc). We evaluated the impact of topical (TOP), subcutaneous (SC), and intraperitoneal (IP) administration of MSC-sEVs on dermal fibrosis, collagen density, and thickness. All three routes of administration significantly reduced BLM-induced fibrosis in the skin, as determined by Masson's Trichrome staining. However, only TOP administration reduced BLM-induced dermal collagen density, with no effect on dermal thickness observed for all administration routes. Moreover, SC, but not TOP or IP administration, increased anti-inflammatory profibrotic CD163+ M2 macrophages. These findings indicate that the administration route influences the therapeutic efficacy of MSC-sEVs in alleviating dermal fibrosis, with TOP administration being the most effective, and this efficacy is not mediated by M2 macrophages. Since both TOP and SC administration target the skin, the difference in their efficacy likely stems from variations in MSC-sEV delivery in the skin. Fluorescence-labelled TOP, but not SC MSC-sEVs when applied to skin explant cultures, localized in the stratum corneum. Hence, the superior efficacy of TOP over SC MSC-sEVs could be attributed to this localization. A comparison of the proteomes of stratum corneum and MSC-sEVs revealed the presence of >100 common proteins. Most of these proteins, such as filaggrin, were known to be crucial for maintaining skin barrier function against irritants and toxins, thereby mitigating inflammation-induced fibrosis. Therefore, the superior efficacy of TOP MSC-sEVs over SC and IP MSC-sEVs against SSc is mediated by the delivery of proteins to the stratum corneum to reinforce the skin barrier.
Collapse
Affiliation(s)
- Bin Zhang
- Paracrine Therapeutics Pte. Ltd., 1 Tai Seng Ave, #02-04 Tai Seng Exchange, Singapore 536464, Singapore; (B.Z.); (R.C.L.); (W.K.S.); (T.T.T.)
| | - Ruenn Chai Lai
- Paracrine Therapeutics Pte. Ltd., 1 Tai Seng Ave, #02-04 Tai Seng Exchange, Singapore 536464, Singapore; (B.Z.); (R.C.L.); (W.K.S.); (T.T.T.)
| | - Wei Kian Sim
- Paracrine Therapeutics Pte. Ltd., 1 Tai Seng Ave, #02-04 Tai Seng Exchange, Singapore 536464, Singapore; (B.Z.); (R.C.L.); (W.K.S.); (T.T.T.)
| | - Thong Teck Tan
- Paracrine Therapeutics Pte. Ltd., 1 Tai Seng Ave, #02-04 Tai Seng Exchange, Singapore 536464, Singapore; (B.Z.); (R.C.L.); (W.K.S.); (T.T.T.)
| | - Sai Kiang Lim
- Paracrine Therapeutics Pte. Ltd., 1 Tai Seng Ave, #02-04 Tai Seng Exchange, Singapore 536464, Singapore; (B.Z.); (R.C.L.); (W.K.S.); (T.T.T.)
- Department of Surgery, YLL School of Medicine, National University Singapore (NUS), 5 Lower Kent Ridge Road, Singapore 119074, Singapore
| |
Collapse
|
5
|
Zhang C, Zhou X, Wang D, Hao L, Zeng Z, Su L. Hydrogel-Loaded Exosomes: A Promising Therapeutic Strategy for Musculoskeletal Disorders. J Clin Pharm Ther 2023; 2023:1-36. [DOI: 10.1155/2023/1105664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/26/2024]
Abstract
Clinical treatment strategies for musculoskeletal disorders have been a hot research topic. Accumulating evidence suggests that hydrogels loaded with MSC-derived EVs show great potential in improving musculoskeletal injuries. The ideal hydrogels should be capable of promoting the development of new tissues and simulating the characteristics of target tissues, with the properties matching the cell-matrix constituents of autologous tissues. Although there have been numerous reports of hydrogels loaded with MSC-derived EVs for the repair of musculoskeletal injuries, such as intervertebral disc injury, tendinopathy, bone fractures, and cartilage injuries, there are still many hurdles to overcome before the clinical application of modified hydrogels. In this review, we focus on the advantages of the isolation technique of EVs in combination with different types of hydrogels. In this context, the efficacy of hydrogels loaded with MSC-derived EVs in different musculoskeletal injuries is discussed in detail to provide a reference for the future application of hydrogels loaded with MSC-derived EVs in the clinical treatment of musculoskeletal injuries.
Collapse
Affiliation(s)
- Chunyu Zhang
- School of Sport Medicine and Rehabilitation, Beijing Sport University, Beijing 100084, China
| | - Xuchang Zhou
- School of Sport Medicine and Rehabilitation, Beijing Sport University, Beijing 100084, China
| | - Dongxue Wang
- School of Sport Medicine and Rehabilitation, Beijing Sport University, Beijing 100084, China
| | - Li Hao
- Shougang Technician College, Nursing School, Beijing 100043, China
- Department of Rehabilitation, Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou 510000, China
| | - Zhipeng Zeng
- School of Sport Medicine and Rehabilitation, Beijing Sport University, Beijing 100084, China
- Shougang Technician College, Nursing School, Beijing 100043, China
- Department of Rehabilitation, Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou 510000, China
| | - Lei Su
- Department of Rehabilitation, Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou 510000, China
| |
Collapse
|
6
|
Nicodemou A, Bernátová S, Čeháková M, Danišovič Ľ. Emerging Roles of Mesenchymal Stem/Stromal-Cell-Derived Extracellular Vesicles in Cancer Therapy. Pharmaceutics 2023; 15:pharmaceutics15051453. [PMID: 37242693 DOI: 10.3390/pharmaceutics15051453] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 04/24/2023] [Accepted: 05/06/2023] [Indexed: 05/28/2023] Open
Abstract
Despite the tremendous efforts of many researchers and clinicians, cancer remains the second leading cause of mortality worldwide. Mesenchymal stem/stromal cells (MSCs) are multipotent cells residing in numerous human tissues and presenting unique biological properties, such as low immunogenicity, powerful immunomodulatory and immunosuppressive capabilities, and, in particular, homing abilities. Therapeutic functions of MSCs are mediated mostly by the paracrine effect of released functional molecules and other variable components, and among them the MSC-derived extracellular vesicles (MSC-EVs) seem to be one of the central mediators of the therapeutic functions of MSCs. MSC-EVs are membrane structures secreted by the MSCs, rich in specific proteins, lipids, and nucleic acids. Amongst these, microRNAs have achieved the most attention currently. Unmodified MSC-EVs can promote or inhibit tumor growth, while modified MSC-EVs are involved in the suppression of cancer progression via the delivery of therapeutic molecules, including miRNAs, specific siRNAs, or suicide RNAs, as well as chemotherapeutic drugs. Here, we present an overview of the characteristics of the MSCs-EVs and describe the current methods for their isolation and analysis, the content of their cargo, and modalities for the modification of MSC-EVs in order for them to be used as drug delivery vehicles. Finally, we describe different roles of MSC-EVs in the tumor microenvironment and summarize current advances of MCS-EVs in cancer research and therapy. MSC-EVs are expected to be a novel and promising cell-free therapeutic drug delivery vehicle for the treatment of cancer.
Collapse
Affiliation(s)
- Andreas Nicodemou
- Lambda Life a. s., Levocska 3617/3, 851 01 Bratislava, Slovakia
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University, Sasinkova 4, 811 08 Bratislava, Slovakia
| | - Soňa Bernátová
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University, Sasinkova 4, 811 08 Bratislava, Slovakia
| | - Michaela Čeháková
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University, Sasinkova 4, 811 08 Bratislava, Slovakia
| | - Ľuboš Danišovič
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University, Sasinkova 4, 811 08 Bratislava, Slovakia
- Centre for Tissue Engineering and Regenerative Medicine-Translational Research Unit in the Branch of Regenerative Medicine, Faculty of Medicine, Comenius University, Bratislava, Sasinkova 4, 811 08 Bratislava, Slovakia
| |
Collapse
|
7
|
Palacio PL, Pleet ML, Reátegui E, Magaña SM. Emerging role of extracellular vesicles in multiple sclerosis: From cellular surrogates to pathogenic mediators and beyond. J Neuroimmunol 2023; 377:578064. [PMID: 36934525 PMCID: PMC10124134 DOI: 10.1016/j.jneuroim.2023.578064] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 02/06/2023] [Accepted: 03/05/2023] [Indexed: 03/16/2023]
Abstract
Multiple Sclerosis (MS) is a chronic, inflammatory demyelinating disease of the central nervous system (CNS) driven by a complex interplay of genetic and environmental factors. While the therapeutic arsenal has expanded significantly for management of relapsing forms of MS, treatment of individuals with progressive MS is suboptimal. This treatment inequality is in part due to an incomplete understanding of pathomechanisms at different stages of the disease-underscoring the critical need for new biomarkers. Extracellular vesicles (EVs) and their bioactive cargo have emerged as endogenous nanoparticles with great theranostic potential-as diagnostic and prognostic biomarkers and ultimately as therapeutic candidates for precision nanotherapeutics. The goals of this review are to: 1) summarize the current data investigating the role of EVs and their bioactive cargo in MS pathogenesis, 2) provide a high level overview of advances and challenges in EV isolation and characterization for translational studies, and 3) conclude with future perspectives on this evolving field.
Collapse
Affiliation(s)
- Paola Loreto Palacio
- Department of Pediatrics, Division of Neurology, Nationwide Children's Hospital, Columbus, OH, USA
| | - Michelle L Pleet
- Viral Immunology Section, Neuroimmunology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Eduardo Reátegui
- William G. Lowrie Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH, USA
| | - Setty M Magaña
- Department of Pediatrics, Division of Neurology, Nationwide Children's Hospital, Columbus, OH, USA.
| |
Collapse
|
8
|
Williams T, Salmanian G, Burns M, Maldonado V, Smith E, Porter RM, Song YH, Samsonraj RM. Versatility of mesenchymal stem cell-derived extracellular vesicles in tissue repair and regenerative applications. Biochimie 2023; 207:33-48. [PMID: 36427681 DOI: 10.1016/j.biochi.2022.11.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 10/29/2022] [Accepted: 11/21/2022] [Indexed: 11/24/2022]
Abstract
Mesenchymal stem/stromal cells (MSCs) are multipotent somatic cells that have been widely explored in the field of regenerative medicine. MSCs possess the ability to secrete soluble factors as well as lipid bound extracellular vesicles (EVs). MSCs have gained increased interest and attention as a result of their therapeutic properties, which are thought to be attributed to their secretome. However, while the use of MSCs as whole cells pose heterogeneity concerns and survival issues post-transplantation, such limitations are absent in cell-free EV-based treatments. EVs derived from MSCs are promising therapeutic agents for a range of clinical conditions and disorders owing to their immunomodulatory, pro-regenerative, anti-inflammatory, and antifibrotic activity. Recent successes with preclinical studies using EVs for repair and regeneration of damaged tissues such as cardiac tissue, lung, liver, pancreas, bone, skin, cornea, and blood diseases are discussed in this review. We also discuss delivery strategies of EVs using biomaterials as delivery vehicles through systemic or local administration. Despite its effectiveness in preclinical investigations, the application of MSC-EV in clinical settings will necessitate careful consideration surrounding issues such as: i) scalability and isolation, ii) biodistribution, iii) targeting specific tissues, iv) quantification and characterization, and v) safety and efficacy of dosage. The future of EVs in regenerative medicine is promising yet still needs further investigation on enhancing the efficacy, scalability, and potency for clinical applications.
Collapse
Affiliation(s)
- Taylor Williams
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, AR, USA
| | - Ghazaleh Salmanian
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, AR, USA
| | - Morgan Burns
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, AR, USA
| | - Vitali Maldonado
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, AR, USA
| | - Emma Smith
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, AR, USA
| | - Ryan M Porter
- Department of Orthopedic Surgery, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Young Hye Song
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, AR, USA; Interdisciplinary Graduate Program in Cell and Molecular Biology, University of Arkansas, Fayetteville, AR, USA
| | - Rebekah Margaret Samsonraj
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, AR, USA; Interdisciplinary Graduate Program in Cell and Molecular Biology, University of Arkansas, Fayetteville, AR, USA; Department of Orthopedic Surgery, University of Arkansas for Medical Sciences, Little Rock, AR, USA.
| |
Collapse
|
9
|
Cortes-Galvez D, Dangerfield JA, Metzner C. Extracellular Vesicles and Their Membranes: Exosomes vs. Virus-Related Particles. MEMBRANES 2023; 13:397. [PMID: 37103824 PMCID: PMC10146078 DOI: 10.3390/membranes13040397] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/28/2023] [Accepted: 03/29/2023] [Indexed: 06/19/2023]
Abstract
Cells produce nanosized lipid membrane-enclosed vesicles which play important roles in intercellular communication. Interestingly, a certain type of extracellular vesicle, termed exosomes, share physical, chemical, and biological properties with enveloped virus particles. To date, most similarities have been discovered with lentiviral particles, however, other virus species also frequently interact with exosomes. In this review, we will take a closer look at the similarities and differences between exosomes and enveloped viral particles, with a focus on events taking place at the vesicle or virus membrane. Since these structures present an area with an opportunity for interaction with target cells, this is relevant for basic biology as well as any potential research or medical applications.
Collapse
Affiliation(s)
- Daniela Cortes-Galvez
- AG Histology and Embryology, Institute of Morphology, University of Veterinary Medicine Vienna, 1210 Vienna, Austria
| | | | | |
Collapse
|
10
|
Zhao H, Li Z, Wang Y, Zhou K, Li H, Bi S, Wang Y, Wu W, Huang Y, Peng B, Tang J, Pan B, Wang B, Chen Z, Zhang Z. Bioengineered MSC-derived exosomes in skin wound repair and regeneration. Front Cell Dev Biol 2023; 11:1029671. [PMID: 36923255 PMCID: PMC10009159 DOI: 10.3389/fcell.2023.1029671] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Accepted: 02/16/2023] [Indexed: 03/02/2023] Open
Abstract
Refractory skin defects such as pressure ulcers, diabetic ulcers, and vascular ulcers represent a challenge for clinicians and researchers in many aspects. The treatment strategies for wound healing have high cost and limited efficacy. To ease the financial and psychological burden on patients, a more effective therapeutic approach is needed to address the chronic wound. MSC-derived exosomes (MSC-exosomes), the main bioactive extracellular vesicles of the paracrine effect of MSCs, have been proposed as a new potential cell-free approach for wound healing and skin regeneration. The benefits of MSC-exosomes include their ability to promote angiogenesis and cell proliferation, increase collagen production, regulate inflammation, and finally improve tissue regenerative capacity. However, poor targeting and easy removability of MSC-exosomes from the wound are major obstacles to their use in clinical therapy. Thus, the concept of bioengineering technology has been introduced to modify exosomes, enabling higher concentrations and construction of particles of greater stability with specific therapeutic capability. The use of biomaterials to load MSC-exosomes may be a promising strategy to concentrate dose, create the desired therapeutic efficacy, and maintain a sustained release effect. The beneficial role of MSC-exosomes in wound healing is been widely accepted; however, the potential of bioengineering-modified MSC-exosomes remains unclear. In this review, we attempt to summarize the therapeutic applications of modified MSC-exosomes in wound healing and skin regeneration. The challenges and prospects of bioengineered MSC-exosomes are also discussed.
Collapse
Affiliation(s)
- Hanxing Zhao
- Department of Plastic and Burn Surgery, West China Hospital, Sichuan University, Chengdu, China.,Department of Plastic Reconstructive and Aesthetic Surgery, West China Tianfu Hospital, Sichuan University, Chengdu, China
| | - Zhengyong Li
- Department of Plastic and Burn Surgery, West China Hospital, Sichuan University, Chengdu, China.,Department of Plastic Reconstructive and Aesthetic Surgery, West China Tianfu Hospital, Sichuan University, Chengdu, China
| | - Yixi Wang
- Department of Plastic and Burn Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Kai Zhou
- Department of Plastic Reconstructive and Aesthetic Surgery, West China Tianfu Hospital, Sichuan University, Chengdu, China
| | - Hairui Li
- Department of Plastic Reconstructive and Aesthetic Surgery, West China Tianfu Hospital, Sichuan University, Chengdu, China
| | - Siwei Bi
- Department of Plastic and Burn Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Yudong Wang
- Department of Plastic and Burn Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Wenqing Wu
- Department of Plastic and Burn Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Yeqian Huang
- Department of Plastic and Burn Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Bo Peng
- Department of Plastic and Burn Surgery, West China Hospital, Sichuan University, Chengdu, China.,Department of Plastic Reconstructive and Aesthetic Surgery, West China Tianfu Hospital, Sichuan University, Chengdu, China
| | - Jun Tang
- Department of Plastic and Burn Surgery, West China Hospital, Sichuan University, Chengdu, China.,Department of Plastic Reconstructive and Aesthetic Surgery, West China Tianfu Hospital, Sichuan University, Chengdu, China
| | - Bo Pan
- Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Baoyun Wang
- Department of Plastic and Burn Surgery, West China Hospital, Sichuan University, Chengdu, China.,Department of Plastic Reconstructive and Aesthetic Surgery, West China Tianfu Hospital, Sichuan University, Chengdu, China
| | - Zhixing Chen
- Department of Plastic and Burn Surgery, West China Hospital, Sichuan University, Chengdu, China.,Department of Plastic Reconstructive and Aesthetic Surgery, West China Tianfu Hospital, Sichuan University, Chengdu, China
| | - Zhenyu Zhang
- Department of Plastic and Burn Surgery, West China Hospital, Sichuan University, Chengdu, China.,Department of Plastic Reconstructive and Aesthetic Surgery, West China Tianfu Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
11
|
Guillamat-Prats R. Role of Mesenchymal Stem/Stromal Cells in Coagulation. Int J Mol Sci 2022; 23:ijms231810393. [PMID: 36142297 PMCID: PMC9499599 DOI: 10.3390/ijms231810393] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 09/02/2022] [Accepted: 09/05/2022] [Indexed: 11/23/2022] Open
Abstract
Mesenchymal stem/stromal cells (MSCs) are widely used in disease models in order to control several phases in the response to injuries, immune reaction, wound healing, and regeneration. MSCs can act upon both the innate and adaptive immune systems and target a broad number of functions, such as the secretion of cytokines, proteolytic enzymes, angiogenic factors, and the regulating of cell proliferation and survival. The role of MSCs in coagulation has been less studied. This review evaluates the properties and main functions of MSCs in coagulation. MSCs can regulate coagulation in a wide range of pathways. MSCs express and release tissue factors (TF), one of the key regulators of the extrinsic coagulation pathways; MSCs can trigger platelet production and contribute to platelet activation. Altogether, MSCs seem to have a pro-thrombotic role and their superior characterization prior to their administration is necessary in order to prevent adverse coagulation events.
Collapse
Affiliation(s)
- Raquel Guillamat-Prats
- Lung Immunity Translational Research Group in Respiratory Diseases, Germans Trias i Pujol Research Institute (IGTP), 08914 Badalona, Spain
| |
Collapse
|
12
|
Pathophysiology of Sepsis and Genesis of Septic Shock: The Critical Role of Mesenchymal Stem Cells (MSCs). Int J Mol Sci 2022; 23:ijms23169274. [PMID: 36012544 PMCID: PMC9409099 DOI: 10.3390/ijms23169274] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 08/14/2022] [Accepted: 08/15/2022] [Indexed: 11/17/2022] Open
Abstract
The treatment of sepsis and septic shock remains a major public health issue due to the associated morbidity and mortality. Despite an improvement in the understanding of the physiological and pathological mechanisms underlying its genesis and a growing number of studies exploring an even higher range of targeted therapies, no significant clinical progress has emerged in the past decade. In this context, mesenchymal stem cells (MSCs) appear more and more as an attractive approach for cell therapy both in experimental and clinical models. Pre-clinical data suggest a cornerstone role of these cells and their secretome in the control of the host immune response. Host-derived factors released from infected cells (i.e., alarmins, HMGB1, ATP, DNA) as well as pathogen-associated molecular patterns (e.g., LPS, peptidoglycans) can activate MSCs located in the parenchyma and around vessels to upregulate the expression of cytokines/chemokines and growth factors that influence, respectively, immune cell recruitment and stem cell mobilization. However, the way in which MSCs exert their beneficial effects in terms of survival and control of inflammation in septic states remains unclear. This review presents the interactions identified between MSCs and mediators of immunity and tissue repair in sepsis. We also propose paradigms related to the plausible roles of MSCs in the process of sepsis and septic shock. Finally, we offer a presentation of experimental and clinical studies and open the way to innovative avenues of research involving MSCs from a prognostic, diagnostic, and therapeutic point of view in sepsis.
Collapse
|
13
|
Soares MBP, Gonçalves RGJ, Vasques JF, da Silva-Junior AJ, Gubert F, Santos GC, de Santana TA, Almeida Sampaio GL, Silva DN, Dominici M, Mendez-Otero R. Current Status of Mesenchymal Stem/Stromal Cells for Treatment of Neurological Diseases. Front Mol Neurosci 2022; 15:883378. [PMID: 35782379 PMCID: PMC9244712 DOI: 10.3389/fnmol.2022.883378] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 05/19/2022] [Indexed: 11/13/2022] Open
Abstract
Neurological disorders include a wide spectrum of clinical conditions affecting the central and peripheral nervous systems. For these conditions, which affect hundreds of millions of people worldwide, generally limited or no treatments are available, and cell-based therapies have been intensively investigated in preclinical and clinical studies. Among the available cell types, mesenchymal stem/stromal cells (MSCs) have been widely studied but as yet no cell-based treatment exists for neurological disease. We review current knowledge of the therapeutic potential of MSC-based therapies for neurological diseases, as well as possible mechanisms of action that may be explored to hasten the development of new and effective treatments. We also discuss the challenges for culture conditions, quality control, and the development of potency tests, aiming to generate more efficient cell therapy products for neurological disorders.
Collapse
Affiliation(s)
- Milena B. P. Soares
- Laboratório de Engenharia Tecidual e Imunofarmacologia, Instituto Gonçalo Moniz, Fundação Oswaldo Cruz (IGM-FIOCRUZ/BA), Salvador, Brazil
- Instituto SENAI de Sistemas Avançados de Saúde (CIMATEC ISI-SAS), Centro Universitário SENAI/CIMATEC, Salvador, Brazil
| | - Renata G. J. Gonçalves
- Laboratório de Neurobiologia Celular e Molecular, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Programa Redes de Pesquisa em Saúde no Estado do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Juliana F. Vasques
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Almir J. da Silva-Junior
- Laboratório de Neurobiologia Celular e Molecular, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Programa Redes de Pesquisa em Nanotecnologia no Estado do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Fernanda Gubert
- Programa Redes de Pesquisa em Saúde no Estado do Rio de Janeiro, Rio de Janeiro, Brazil
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Girlaine Café Santos
- Laboratório de Engenharia Tecidual e Imunofarmacologia, Instituto Gonçalo Moniz, Fundação Oswaldo Cruz (IGM-FIOCRUZ/BA), Salvador, Brazil
- Instituto SENAI de Sistemas Avançados de Saúde (CIMATEC ISI-SAS), Centro Universitário SENAI/CIMATEC, Salvador, Brazil
| | - Thaís Alves de Santana
- Laboratório de Engenharia Tecidual e Imunofarmacologia, Instituto Gonçalo Moniz, Fundação Oswaldo Cruz (IGM-FIOCRUZ/BA), Salvador, Brazil
- Instituto SENAI de Sistemas Avançados de Saúde (CIMATEC ISI-SAS), Centro Universitário SENAI/CIMATEC, Salvador, Brazil
| | - Gabriela Louise Almeida Sampaio
- Laboratório de Engenharia Tecidual e Imunofarmacologia, Instituto Gonçalo Moniz, Fundação Oswaldo Cruz (IGM-FIOCRUZ/BA), Salvador, Brazil
- Instituto SENAI de Sistemas Avançados de Saúde (CIMATEC ISI-SAS), Centro Universitário SENAI/CIMATEC, Salvador, Brazil
| | | | - Massimo Dominici
- Laboratory of Cellular Therapy, Division of Oncology, University of Modena and Reggio Emilia (UNIMORE), Modena, Italy
| | - Rosalia Mendez-Otero
- Laboratório de Neurobiologia Celular e Molecular, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Programa Redes de Pesquisa em Saúde no Estado do Rio de Janeiro, Rio de Janeiro, Brazil
- Programa Redes de Pesquisa em Nanotecnologia no Estado do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
14
|
Yue C, Cao J, Wong A, Kim JH, Alam S, Luong G, Talegaonkar S, Schwartz Z, Boyan BD, Giannobile WV, Sahingur SE, Lin Z. Human Bone Marrow Stromal Cell Exosomes Ameliorate Periodontitis. J Dent Res 2022; 101:1110-1118. [PMID: 35356822 PMCID: PMC9305845 DOI: 10.1177/00220345221084975] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Human bone marrow stromal cell (hBMSC)-derived exosomes are promising therapeutics for inflammatory diseases due to their unique microRNA (miRNA) and protein cargos. Periodontal diseases often present with chronicity and corresponding exuberant inflammation, which leads to loss of tooth support. In this study, we explored whether hBMSC exosomes can affect periodontitis progression. hBMSC exosomes were isolated from cell culture medium through sequential ultracentrifugation. miRNAs and proteins that were enriched in hBMSC exosomes were characterized by RNA sequencing and protein array, respectively. hBMSC exosomes significantly suppressed periodontal keystone pathogen Porphyromonas gingivalis-triggered inflammatory response in macrophages in vitro. Transcriptomic analysis suggested that exosomes exerted their effects through regulating cell metabolism, differentiation, and inflammation resolution. In vivo, weekly exosome injection into the gingival tissues reduced the tissue destruction and immune cell infiltration in rat ligature-induced periodontitis model. Collectively, these findings suggest that hBMSC-derived exosomes can potentially be used as a host modulation agent in the management of periodontitis.
Collapse
Affiliation(s)
- C Yue
- Department of Periodontics, School of Dentistry, Virginia Commonwealth University, Richmond, VA, USA
| | - J Cao
- Department of Periodontics, School of Dentistry, Virginia Commonwealth University, Richmond, VA, USA.,Department of Periodontology, Peking University School and Hospital of Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Beijing, People's Republic of China
| | - A Wong
- Department of Periodontics, School of Dentistry, Virginia Commonwealth University, Richmond, VA, USA
| | - J H Kim
- Department of Periodontics, School of Dentistry, Virginia Commonwealth University, Richmond, VA, USA
| | - S Alam
- Department of Periodontics, School of Dentistry, Virginia Commonwealth University, Richmond, VA, USA
| | - G Luong
- Department of Periodontics, School of Dentistry, Virginia Commonwealth University, Richmond, VA, USA
| | - S Talegaonkar
- Department of Periodontics, School of Dentistry, Virginia Commonwealth University, Richmond, VA, USA
| | - Z Schwartz
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA, USA
| | - B D Boyan
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA, USA
| | - W V Giannobile
- Department of Oral Medicine, Infection, and Immunity, Harvard School of Dental Medicine, Boston, MA, USA
| | - S E Sahingur
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, PA, USA
| | - Z Lin
- Department of Periodontics, School of Dentistry, Virginia Commonwealth University, Richmond, VA, USA
| |
Collapse
|
15
|
Landfield Q, Saito M, Hashim A, Canals-Baker S, Sershen H, Levy E, Saito M. Cocaine Induces Sex-Associated Changes in Lipid Profiles of Brain Extracellular Vesicles. Neurochem Res 2021; 46:2909-2922. [PMID: 34245421 PMCID: PMC8490334 DOI: 10.1007/s11064-021-03395-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 07/01/2021] [Accepted: 07/03/2021] [Indexed: 11/27/2022]
Abstract
Cocaine is a highly addictive stimulant with diverse effects on physiology. Recent studies indicate the involvement of extracellular vesicles (EVs) secreted by neural cells in the cocaine addiction process. It is hypothesized that cocaine affects secretion levels of EVs and their cargos, resulting in modulation of synaptic transmission and plasticity related to addiction physiology and pathology. Lipids present in EVs are important for EV formation and for intercellular lipid exchange that may trigger physiological and pathological responses, including neuroplasticity, neurotoxicity, and neuroinflammation. Specific lipids are highly enriched in EVs compared to parent cells, and recent studies suggest the involvement of various lipids in drug-induced synaptic plasticity during the development and maintenance of addiction processes. Therefore, we examined interstitial small EVs isolated from the brain of mice treated with either saline or cocaine, focusing on the effects of cocaine on the lipid composition of EVs. We demonstrate that 12 days of noncontingent repeated cocaine (10 mg/kg) injections to mice, which induce locomotor sensitization, cause lipid composition changes in brain EVs of male mice as compared with saline-injected controls. The most prominent change is the elevation of GD1a ganglioside in brain EVs of males. However, cocaine does not affect the EV lipid profiles of the brain in female mice. Understanding the relationship between lipid composition in EVs and vulnerability to cocaine addiction may provide insight into novel targets for therapies for addiction.
Collapse
Affiliation(s)
- Qwynn Landfield
- Division of Neurochemistry, Nathan S. Kline Institute for Psychiatric Research, 140 Old Orangeburg Rd., Orangeburg, NY, 10962, USA
| | - Mitsuo Saito
- Division of Neurochemistry, Nathan S. Kline Institute for Psychiatric Research, 140 Old Orangeburg Rd., Orangeburg, NY, 10962, USA
| | - Audrey Hashim
- Division of Neurochemistry, Nathan S. Kline Institute for Psychiatric Research, 140 Old Orangeburg Rd., Orangeburg, NY, 10962, USA
| | - Stefanie Canals-Baker
- Division of Neurochemistry, Nathan S. Kline Institute for Psychiatric Research, 140 Old Orangeburg Rd., Orangeburg, NY, 10962, USA
| | - Henry Sershen
- Division of Neurochemistry, Nathan S. Kline Institute for Psychiatric Research, 140 Old Orangeburg Rd., Orangeburg, NY, 10962, USA
- Department of Psychiatry, New York University School of Medicine, New York, NY, USA
| | - Efrat Levy
- Department of Psychiatry, New York University School of Medicine, New York, NY, USA
- Center for Dementia Research, Nathan S. Kline Institute for Psychiatric Research, Orangeburg, NY, USA
- NYU Neuroscience Institute, New York University School of Medicine, New York, NY, USA
- Department of Biochemistry & Molecular Pharmacology, New York University School of Medicine, New York, NY, USA
| | - Mariko Saito
- Division of Neurochemistry, Nathan S. Kline Institute for Psychiatric Research, 140 Old Orangeburg Rd., Orangeburg, NY, 10962, USA.
- Department of Psychiatry, New York University School of Medicine, New York, NY, USA.
| |
Collapse
|
16
|
Pathipati P, Lecuyer M, Faustino J, Strivelli J, Phinney DG, Vexler ZS. Mesenchymal Stem Cell (MSC)-Derived Extracellular Vesicles Protect from Neonatal Stroke by Interacting with Microglial Cells. Neurotherapeutics 2021; 18:1939-1952. [PMID: 34235636 PMCID: PMC8609070 DOI: 10.1007/s13311-021-01076-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/16/2021] [Indexed: 02/04/2023] Open
Abstract
Mesenchymal stem cell (MSC)-based therapies are beneficial in models of perinatal stroke and hypoxia-ischemia. Mounting evidence suggests that in adult injury models, including stroke, MSC-derived small extracellular vesicles (MSC-sEV) contribute to the neuroprotective and regenerative effects of MSCs. Herein, we examined if MSC-sEV protect neonatal brain from stroke and if this effect is mediated via communication with microglia. MSC-sEV derived from bone marrow MSCs were characterized by size distribution (NanoSight™) and identity (protein markers). Studies in microglial cells isolated from the injured or contralateral cortex of postnatal day 9 (P9) mice subjected to a 3-h middle cerebral artery occlusion (tMCAO) and cultured (in vitro) revealed that uptake of fluorescently labeled MSC-sEV was significantly greater by microglia from the injured cortex vs. contralateral cortex. The cell-type-specific spatiotemporal distribution of MSC-sEV was also determined in vivo after tMCAO at P9. MSC-sEV administered at reperfusion, either by intracerebroventricular (ICV) or by intranasal (IN) routes, accumulated in the hemisphere ipsilateral to the occlusion, with differing spatial distribution 2 h, 18 h, and 72 h regardless of the administration route. By 72 h, MSC-sEV in the IN group was predominantly observed in Iba1+ cells with retracted processes and in GLUT1+ blood vessels in ischemic-reperfused regions. MSC-sEV presence in Iba1+ cells was sustained. MSC-sEV administration also significantly reduced injury volume 72 h after tMCAO in part via modulatory effects on microglial cells. Together, these data establish feasibility for MSC-sEV delivery to injured neonatal brain via a clinically relevant IN route, which affords protection during sub-acute injury phase.
Collapse
Affiliation(s)
- Praneeti Pathipati
- Department of Neurology, UCSF, 675 Nelson Rising Lane, San Francisco, CA, 94158-0663, USA
- Department of Pediatrics, UCSF, San Francisco, CA, USA
| | - Matthieu Lecuyer
- Department of Neurology, UCSF, 675 Nelson Rising Lane, San Francisco, CA, 94158-0663, USA
| | - Joel Faustino
- Department of Neurology, UCSF, 675 Nelson Rising Lane, San Francisco, CA, 94158-0663, USA
| | | | - Donald G Phinney
- Department of Molecular Medicine, Scripps Research Institute, Jupiter, FL, USA
| | - Zinaida S Vexler
- Department of Neurology, UCSF, 675 Nelson Rising Lane, San Francisco, CA, 94158-0663, USA.
| |
Collapse
|
17
|
Donoso‐Quezada J, Ayala‐Mar S, González‐Valdez J. The role of lipids in exosome biology and intercellular communication: Function, analytics and applications. Traffic 2021; 22:204-220. [PMID: 34053166 PMCID: PMC8361711 DOI: 10.1111/tra.12803] [Citation(s) in RCA: 128] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Revised: 05/18/2021] [Accepted: 05/27/2021] [Indexed: 12/16/2022]
Abstract
Exosomes are extracellular vesicles that in recent years have received special attention for their regulatory functions in numerous biological processes. Recent evidence suggests a correlation between the composition of exosomes in body fluids and the progression of some disorders, such as cancer, diabetes and neurodegenerative diseases. In consequence, numerous studies have been performed to evaluate the composition of these vesicles, aiming to develop new biomarkers for diagnosis and to find novel therapeutic targets. On their part, lipids represent one of the most important components of exosomes, with important structural and regulatory functions during exosome biogenesis, release, targeting and cellular uptake. Therefore, exosome lipidomics has emerged as an innovative discipline for the discovery of novel lipid species with biomedical applications. This review summarizes the current knowledge about exosome lipids and their roles in exosome biology and intercellular communication. Furthermore, it presents the state-of-the-art analytical procedures used in exosome lipidomics while emphasizing how this emerging discipline is providing new insights for future applications of exosome lipids in biomedicine.
Collapse
Affiliation(s)
| | - Sergio Ayala‐Mar
- Tecnologico de MonterreySchool of Engineering and ScienceMonterreyNuevo LeónMexico
| | - José González‐Valdez
- Tecnologico de MonterreySchool of Engineering and ScienceMonterreyNuevo LeónMexico
| |
Collapse
|
18
|
Pils V, Terlecki-Zaniewicz L, Schosserer M, Grillari J, Lämmermann I. The role of lipid-based signalling in wound healing and senescence. Mech Ageing Dev 2021; 198:111527. [PMID: 34174292 DOI: 10.1016/j.mad.2021.111527] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 05/28/2021] [Accepted: 06/17/2021] [Indexed: 02/07/2023]
Abstract
Lipid-based signalling modulates several cellular processes and intercellular communication during wound healing and tissue regeneration. Bioactive lipids include but are not limited to the diverse group of eicosanoids, phospholipids, and extracellular vesicles and mediate the attraction of immune cells, initiation of inflammatory responses, and their resolution. In aged individuals, wound healing and tissue regeneration are greatly impaired, resulting in a delayed healing process and non-healing wounds. Senescent cells accumulate with age in vivo, preferably at sites implicated in age-associated pathologies and their elimination was shown to alleviate many age-associated diseases and disorders. In contrast to these findings, the transient presence of senescent cells in the process of wound healing exerts beneficial effects and limits fibrosis. Hence, clearance of senescent cells during wound healing was repeatedly shown to delay wound closure in vivo. Recent findings established a dysregulated synthesis of eicosanoids, phospholipids and extracellular vesicles as part of the senescent phenotype. This intriguing connection between cellular senescence, lipid-based signalling, and the process of wound healing and tissue regeneration prompts us to compile the current knowledge in this review and propose future directions for investigation.
Collapse
Affiliation(s)
- Vera Pils
- Christian Doppler Laboratory for the Biotechnology of Skin Aging, Institute of Molecular Biotechnology, University of Natural Resources and Life Sciences, Vienna, Austria
| | - Lucia Terlecki-Zaniewicz
- Christian Doppler Laboratory for the Biotechnology of Skin Aging, Institute of Molecular Biotechnology, University of Natural Resources and Life Sciences, Vienna, Austria
| | - Markus Schosserer
- Christian Doppler Laboratory for Skin Multimodal Imaging of Aging and Senescence - SKINMAGINE, Institute of Molecular Biotechnology, University of Natural Resources and Life Sciences, Vienna, Austria; Austrian Cluster for Tissue Regeneration, Austria
| | - Johannes Grillari
- Christian Doppler Laboratory for the Biotechnology of Skin Aging, Institute of Molecular Biotechnology, University of Natural Resources and Life Sciences, Vienna, Austria; Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, AUVA Research Center, Linz and Vienna, Austria; Austrian Cluster for Tissue Regeneration, Austria
| | - Ingo Lämmermann
- Christian Doppler Laboratory for the Biotechnology of Skin Aging, Institute of Molecular Biotechnology, University of Natural Resources and Life Sciences, Vienna, Austria.
| |
Collapse
|
19
|
Bryk M, Karnas E, Mlost J, Zuba-Surma E, Starowicz K. Mesenchymal stem cells and extracellular vesicles for the treatment of pain: Current status and perspectives. Br J Pharmacol 2021; 179:4281-4299. [PMID: 34028798 DOI: 10.1111/bph.15569] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 03/26/2021] [Accepted: 05/05/2021] [Indexed: 12/20/2022] Open
Abstract
Mesenchymal stem/stromal cells (MSCs) are multipotent progenitor cells of mesodermal origin. Due to their capacity for self-renewal and differentiation into several cell types, MSCs have been extensively studied in experimental biology and regenerative medicine in recent years. Moreover, MSCs release extracellular vesicles (EVs), which might be partly responsible for their regenerative properties. MSCs regulate several processes in target cells via paracrine signalling, such as immunomodulation, anti-apoptotic signalling, tissue remodelling, angiogenesis and anti-fibrotic signalling. The aim of this review is to provide a detailed description of the functional properties of MSCs and EVs and their potential clinical applications, with a special focus on pain treatment. The analgesic, anti-inflammatory and regenerative properties of MSCs and EVs will be discussed for several diseases, such as neuropathic pain, osteoarthritis and spinal cord injury.
Collapse
Affiliation(s)
- Marta Bryk
- Maj Institute of Pharmacology, Polish Academy of Sciences, Kraków, Poland
| | - Elżbieta Karnas
- Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Jakub Mlost
- Maj Institute of Pharmacology, Polish Academy of Sciences, Kraków, Poland
| | - Ewa Zuba-Surma
- Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | | |
Collapse
|
20
|
Nazari-Shafti TZ, Neuber S, Falk V, Emmert MY. Toward next-generation advanced therapies: extracellular vesicles and cell therapy - partners or competitors? Regen Med 2021; 16:215-218. [PMID: 33622051 DOI: 10.2217/rme-2020-0138] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Affiliation(s)
- Timo Z Nazari-Shafti
- Department of Cardiothoracic and Vascular Surgery, German Heart Center Berlin, 13353 Berlin, Germany.,German Centre for Cardiovascular Research, Partner Site Berlin, 13353 Berlin, Germany.,Berlin Institute of Health Center for Regenerative Therapies, Charité - Universitätsmedizin Berlin, 13353 Berlin, Germany
| | - Sebastian Neuber
- Department of Cardiothoracic and Vascular Surgery, German Heart Center Berlin, 13353 Berlin, Germany.,German Centre for Cardiovascular Research, Partner Site Berlin, 13353 Berlin, Germany.,Berlin Institute of Health Center for Regenerative Therapies, Charité - Universitätsmedizin Berlin, 13353 Berlin, Germany
| | - Volkmar Falk
- Department of Cardiothoracic and Vascular Surgery, German Heart Center Berlin, 13353 Berlin, Germany.,German Centre for Cardiovascular Research, Partner Site Berlin, 13353 Berlin, Germany.,Department of Health Sciences and Technology, ETH Zürich, 8093 Zürich, Switzerland.,Clinic for Cardiovascular Surgery, Charité - Universitätsmedizin Berlin, 13353 Berlin, Germany
| | - Maximilian Y Emmert
- Department of Cardiothoracic and Vascular Surgery, German Heart Center Berlin, 13353 Berlin, Germany.,German Centre for Cardiovascular Research, Partner Site Berlin, 13353 Berlin, Germany.,Berlin Institute of Health Center for Regenerative Therapies, Charité - Universitätsmedizin Berlin, 13353 Berlin, Germany.,Institute for Regenerative Medicine, University of Zürich, 8044 Zürich, Switzerland.,Wyss Zürich, University of Zürich and ETH Zürich, 8092 Zürich, Switzerland
| |
Collapse
|
21
|
Hermann DM, Doeppner TR, Giebel B. Circulating MicroRNAs: Posttranscriptional Regulators and Disease Markers Holding Promise in Stroke Prediction. Stroke 2021; 52:954-956. [PMID: 33563013 DOI: 10.1161/strokeaha.120.033688] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Dirk M Hermann
- Department of Neurology, University Hospital Essen, University of Duisburg-Essen, Germany (D.M.H.)
| | - Thorsten R Doeppner
- Department of Neurology, University Medicine Göttingen, University of Göttingen, Germany (T.R.D.)
| | - Bernd Giebel
- Institute of Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, Germany (B.G.)
| |
Collapse
|
22
|
Adlerz K, Patel D, Rowley J, Ng K, Ahsan T. Strategies for scalable manufacturing and translation of MSC-derived extracellular vesicles. Stem Cell Res 2020; 48:101978. [PMID: 32947235 DOI: 10.1016/j.scr.2020.101978] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 06/25/2020] [Accepted: 08/26/2020] [Indexed: 12/17/2022] Open
Abstract
Mesenchymal Stem/Stromal Cells (MSCs) are a well-studied cellular therapy with many clinical trials over the last few decades to treat a range of therapeutic indications. Recently, extracellular vesicles secreted by MSCs (MSC-EVs) have been shown to recapitulate many of the therapeutic effects of the MSCs themselves. While research in MSC-EVs has exploded, it is still early in their development towards a clinical therapy. One of the main challenges in cellular therapy, which will clearly also be a challenge in MSC-EV manufacturing, is developing a scalable, cGMP-compatible manufacturing paradigm. Therefore, the focus of this review is to identify some key MSC-EV manufacturing considerations such as the selection of critical raw materials, manufacturing platforms, and critical quality attribute assays. Addressing these issues early in research and development will accelerate clinical product development, clinical trials, and commercial therapies of MSC-EVs.
Collapse
Affiliation(s)
- Katrina Adlerz
- RoosterBio, Inc. 5295 Westview Drive, Suite 275, Frederick, MD 21703, USA
| | - Divya Patel
- RoosterBio, Inc. 5295 Westview Drive, Suite 275, Frederick, MD 21703, USA
| | - Jon Rowley
- RoosterBio, Inc. 5295 Westview Drive, Suite 275, Frederick, MD 21703, USA
| | - Kelvin Ng
- Bioprocessing Technology Institute, 20 Biopolis Way, Centros #06-01 138668, Singapore.
| | - Tabassum Ahsan
- RoosterBio, Inc. 5295 Westview Drive, Suite 275, Frederick, MD 21703, USA.
| |
Collapse
|
23
|
Carvalho PHPR, Correa JR, Paiva KLR, Machado DFS, Scholten JD, Neto BAD. Plasma membrane imaging with a fluorescent benzothiadiazole derivative. Beilstein J Org Chem 2019; 15:2644-2654. [PMID: 31807199 PMCID: PMC6880836 DOI: 10.3762/bjoc.15.257] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 10/15/2019] [Indexed: 12/11/2022] Open
Abstract
This work describes a novel fluorescent 2,1,3-benzothiadiazole derivative designed to act as a water-soluble and selective bioprobe for plasma membrane imaging. The new compound was efficiently synthesized in a two-step procedure with good yields. The photophysical properties were evaluated and the dye proved to have an excellent photostability in several solvents. DFT calculations were found in agreement with the experimental data and helped to understand the stabilizing intramolecular charge-transfer process from the first excited state. The new fluorescent derivative could be applied as selective bioprobe in several cell lines and displayed plasma-membrane affinity during the imaging experiments for all tested models.
Collapse
Affiliation(s)
- Pedro H P R Carvalho
- Laboratory of Medicinal and Technological Chemistry, University of Brasília, Chemistry Institute (IQ-UnB), Campus Universitário Darcy Ribeiro, Brasília, Distrito Federal, 70904-970, Brazil
- Laboratory of Molecular Catalysis, Institute of Chemistry, Graduate Program (PPGQ), Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, 91501-970, Brazil
| | - Jose R Correa
- Laboratory of Medicinal and Technological Chemistry, University of Brasília, Chemistry Institute (IQ-UnB), Campus Universitário Darcy Ribeiro, Brasília, Distrito Federal, 70904-970, Brazil
| | - Karen L R Paiva
- Laboratory of Medicinal and Technological Chemistry, University of Brasília, Chemistry Institute (IQ-UnB), Campus Universitário Darcy Ribeiro, Brasília, Distrito Federal, 70904-970, Brazil
| | - Daniel F S Machado
- Laboratory of Medicinal and Technological Chemistry, University of Brasília, Chemistry Institute (IQ-UnB), Campus Universitário Darcy Ribeiro, Brasília, Distrito Federal, 70904-970, Brazil
| | - Jackson D Scholten
- Laboratory of Molecular Catalysis, Institute of Chemistry, Graduate Program (PPGQ), Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, 91501-970, Brazil
| | - Brenno A D Neto
- Laboratory of Medicinal and Technological Chemistry, University of Brasília, Chemistry Institute (IQ-UnB), Campus Universitário Darcy Ribeiro, Brasília, Distrito Federal, 70904-970, Brazil
- Laboratory of Molecular Catalysis, Institute of Chemistry, Graduate Program (PPGQ), Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, 91501-970, Brazil
| |
Collapse
|
24
|
Witwer KW, Van Balkom BW, Bruno S, Choo A, Dominici M, Gimona M, Hill AF, De Kleijn D, Koh M, Lai RC, Mitsialis SA, Ortiz LA, Rohde E, Asada T, Toh WS, Weiss DJ, Zheng L, Giebel B, Lim SK. Defining mesenchymal stromal cell (MSC)-derived small extracellular vesicles for therapeutic applications. J Extracell Vesicles 2019; 8:1609206. [PMID: 31069028 PMCID: PMC6493293 DOI: 10.1080/20013078.2019.1609206] [Citation(s) in RCA: 408] [Impact Index Per Article: 68.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 04/09/2019] [Accepted: 04/14/2019] [Indexed: 12/13/2022] Open
Abstract
Small extracellular vesicles (sEVs) from mesenchymal stromal/stem cells (MSCs) are transiting rapidly towards clinical applications. However, discrepancies and controversies about the biology, functions, and potency of MSC-sEVs have arisen due to several factors: the diversity of MSCs and their preparation; various methods of sEV production and separation; a lack of standardized quality assurance assays; and limited reproducibility of in vitro and in vivo functional assays. To address these issues, members of four societies (SOCRATES, ISEV, ISCT and ISBT) propose specific harmonization criteria for MSC-sEVs to facilitate data sharing and comparison, which should help to advance the field towards clinical applications. Specifically, MSC-sEVs should be defined by quantifiable metrics to identify the cellular origin of the sEVs in a preparation, presence of lipid-membrane vesicles, and the degree of physical and biochemical integrity of the vesicles. For practical purposes, new MSC-sEV preparations might also be measured against a well-characterized MSC-sEV biological reference. The ultimate goal of developing these metrics is to map aspects of MSC-sEV biology and therapeutic potency onto quantifiable features of each preparation.
Collapse
Affiliation(s)
- Kenneth W. Witwer
- Department of Molecular and Comparative Pathobiology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Bas W.M. Van Balkom
- Division Internal Medicine and Dermatology, Nephrology and Hypertension, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Stefania Bruno
- Department of Medical Sciences and Molecular Biotechnology Center, University of Torino, Torino, Italy
| | - Andre Choo
- Bioprocessing Technology Institute, Agency for Science, Technology and Research, Singapore, Singapore
| | - Massimo Dominici
- MAB Laboratory, TPM of Mirandola, Mirandola, Italy
- Division of Oncology, University of Modena and Reggio Emilia, Modena, Italy
| | - Mario Gimona
- GMP Laboratory, Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Research Program Nanovesicular Therapies, Department of Transfusion Medicine and Celericon Therapeutics G.m.b.H., Paracelsus Medical University (PMU), Salzburg, Austria
| | - Andrew F. Hill
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Australia
| | - Dominique De Kleijn
- Department of Vascular Surgery, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Mickey Koh
- Department of Haematology, St George’s University Hospital NHS Trust, London, UK
- Cell Therapy Facility, Blood Services Group Health Sciences Authority, Singapore, Singapore
| | - Ruenn Chai Lai
- Institute of Medical Biology, Agency for Science, Technology and Research, Singapore, Singapore
| | - S. Alex Mitsialis
- Department of Pediatrics, Harvard Medical School & Division of Newborn Medicine, Boston Children’s Hospital, Boston, MA, USA
| | - Luis A. Ortiz
- Division of Environmental and Occupational Medicine, Department of Environmental and Occupational Health, Graduate School of Public Health at the University of Pittsburgh, Pittsburgh, PA, USA
| | - Eva Rohde
- GMP Laboratory, Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Research Program Nanovesicular Therapies, Department of Transfusion Medicine and Celericon Therapeutics G.m.b.H., Paracelsus Medical University (PMU), Salzburg, Austria
| | - Takashi Asada
- Department of Tissue Regeneration Science and Engineering, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Wei Seong Toh
- Faculty of Dentistry, National University of Singapore, Singapore, Singapore
| | - Daniel J. Weiss
- Health Sciences Research Facility, University of Vermont College of Medicine, Burlington, VT, USA
| | - Lei Zheng
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Bernd Giebel
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Sai Kiang Lim
- Institute of Medical Biology, Agency for Science, Technology and Research, Singapore, Singapore
- Department of Surgery, YLL School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
25
|
Silachev DN, Goryunov KV, Shpilyuk MA, Beznoschenko OS, Morozova NY, Kraevaya EE, Popkov VA, Pevzner IB, Zorova LD, Evtushenko EA, Starodubtseva NL, Kononikhin AS, Bugrova AE, Evtushenko EG, Plotnikov EY, Zorov DB, Sukhikh GT. Effect of MSCs and MSC-Derived Extracellular Vesicles on Human Blood Coagulation. Cells 2019; 8:cells8030258. [PMID: 30893822 PMCID: PMC6468445 DOI: 10.3390/cells8030258] [Citation(s) in RCA: 95] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2019] [Revised: 03/09/2019] [Accepted: 03/15/2019] [Indexed: 01/10/2023] Open
Abstract
Mesenchymal stem cells (MSCs) have emerged as a potent therapeutic tool for the treatment of a number of pathologies, including immune pathologies. However, unwelcome effects of MSCs on blood coagulation have been reported, motivating us to explore the thrombotic properties of human MSCs from the umbilical cord. We revealed strong procoagulant effects of MSCs on human blood and platelet-free plasma using rotational thromboelastometry and thrombodynamic tests. A similar potentiation of clotting was demonstrated for MSC-derived extracellular vesicles (EVs). To offer approaches to avoid unwanted effects, we studied the impact of a heparin supplement on MSC procoagulative properties. However, MSCs still retained procoagulant activity toward blood from children receiving a therapeutic dose of unfractionated heparin. An analysis of the mechanisms responsible for the procoagulant effect of MSCs/EVs revealed the presence of tissue factor and other proteins involved in coagulation-associated pathways. Also, we found that some MSCs and EVs were positive for annexin V, which implies the presence of phosphatidylserine on their surfaces, which can potentiate clot formation. Thus, we revealed procoagulant activity of MSCs/EVs associated with the presence of phosphatidylserine and tissue factor, which requires further analysis to avoid adverse effects of MSC therapy in patients with a risk of thrombosis.
Collapse
Affiliation(s)
- Denis N. Silachev
- V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, Moscow 117198, Russia; (D.N.S.); (K.V.G.); (M.A.S.); (N.Y.M.); (E.E.K.); (V.A.P.); (I.B.P.); (L.D.Z.); (N.L.S.); (A.S.K.); (A.E.B.); (G.T.S.)
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119992, Russia
| | - Kirill V. Goryunov
- V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, Moscow 117198, Russia; (D.N.S.); (K.V.G.); (M.A.S.); (N.Y.M.); (E.E.K.); (V.A.P.); (I.B.P.); (L.D.Z.); (N.L.S.); (A.S.K.); (A.E.B.); (G.T.S.)
| | - Margarita A. Shpilyuk
- V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, Moscow 117198, Russia; (D.N.S.); (K.V.G.); (M.A.S.); (N.Y.M.); (E.E.K.); (V.A.P.); (I.B.P.); (L.D.Z.); (N.L.S.); (A.S.K.); (A.E.B.); (G.T.S.)
| | - Olga S. Beznoschenko
- V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, Moscow 117198, Russia; (D.N.S.); (K.V.G.); (M.A.S.); (N.Y.M.); (E.E.K.); (V.A.P.); (I.B.P.); (L.D.Z.); (N.L.S.); (A.S.K.); (A.E.B.); (G.T.S.)
| | - Natalya Y. Morozova
- V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, Moscow 117198, Russia; (D.N.S.); (K.V.G.); (M.A.S.); (N.Y.M.); (E.E.K.); (V.A.P.); (I.B.P.); (L.D.Z.); (N.L.S.); (A.S.K.); (A.E.B.); (G.T.S.)
| | - Elizaveta E. Kraevaya
- V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, Moscow 117198, Russia; (D.N.S.); (K.V.G.); (M.A.S.); (N.Y.M.); (E.E.K.); (V.A.P.); (I.B.P.); (L.D.Z.); (N.L.S.); (A.S.K.); (A.E.B.); (G.T.S.)
| | - Vasily A. Popkov
- V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, Moscow 117198, Russia; (D.N.S.); (K.V.G.); (M.A.S.); (N.Y.M.); (E.E.K.); (V.A.P.); (I.B.P.); (L.D.Z.); (N.L.S.); (A.S.K.); (A.E.B.); (G.T.S.)
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119992, Russia
| | - Irina B. Pevzner
- V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, Moscow 117198, Russia; (D.N.S.); (K.V.G.); (M.A.S.); (N.Y.M.); (E.E.K.); (V.A.P.); (I.B.P.); (L.D.Z.); (N.L.S.); (A.S.K.); (A.E.B.); (G.T.S.)
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119992, Russia
| | - Ljubava D. Zorova
- V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, Moscow 117198, Russia; (D.N.S.); (K.V.G.); (M.A.S.); (N.Y.M.); (E.E.K.); (V.A.P.); (I.B.P.); (L.D.Z.); (N.L.S.); (A.S.K.); (A.E.B.); (G.T.S.)
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119992, Russia
| | | | - Natalia L. Starodubtseva
- V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, Moscow 117198, Russia; (D.N.S.); (K.V.G.); (M.A.S.); (N.Y.M.); (E.E.K.); (V.A.P.); (I.B.P.); (L.D.Z.); (N.L.S.); (A.S.K.); (A.E.B.); (G.T.S.)
- Moscow Institute of Physics and Technology, Moscow 141701, Russia
| | - Alexey S. Kononikhin
- V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, Moscow 117198, Russia; (D.N.S.); (K.V.G.); (M.A.S.); (N.Y.M.); (E.E.K.); (V.A.P.); (I.B.P.); (L.D.Z.); (N.L.S.); (A.S.K.); (A.E.B.); (G.T.S.)
- Moscow Institute of Physics and Technology, Moscow 141701, Russia
| | - Anna E. Bugrova
- V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, Moscow 117198, Russia; (D.N.S.); (K.V.G.); (M.A.S.); (N.Y.M.); (E.E.K.); (V.A.P.); (I.B.P.); (L.D.Z.); (N.L.S.); (A.S.K.); (A.E.B.); (G.T.S.)
- Emanuel Institute of Biochemical Physics, Russian Academy of Sciences, Moscow 119334, Russia
| | | | - Egor Y. Plotnikov
- V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, Moscow 117198, Russia; (D.N.S.); (K.V.G.); (M.A.S.); (N.Y.M.); (E.E.K.); (V.A.P.); (I.B.P.); (L.D.Z.); (N.L.S.); (A.S.K.); (A.E.B.); (G.T.S.)
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119992, Russia
- Correspondence: (E.Y.P.); (D.B.Z.); Tel.: +7-495-939-5944 (E.Y.P. & D.B.Z.)
| | - Dmitry B. Zorov
- V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, Moscow 117198, Russia; (D.N.S.); (K.V.G.); (M.A.S.); (N.Y.M.); (E.E.K.); (V.A.P.); (I.B.P.); (L.D.Z.); (N.L.S.); (A.S.K.); (A.E.B.); (G.T.S.)
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119992, Russia
- Correspondence: (E.Y.P.); (D.B.Z.); Tel.: +7-495-939-5944 (E.Y.P. & D.B.Z.)
| | - Gennady T. Sukhikh
- V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, Moscow 117198, Russia; (D.N.S.); (K.V.G.); (M.A.S.); (N.Y.M.); (E.E.K.); (V.A.P.); (I.B.P.); (L.D.Z.); (N.L.S.); (A.S.K.); (A.E.B.); (G.T.S.)
- Department of Obstetrics, Gynecology, Perinatology and Reproductology, Institute of Professional Education, First Moscow State Medical University Named after I.M. Sechenov, Moscow 119992, Russia
| |
Collapse
|