1
|
Le Lay S, Scherer PE. Exploring adipose tissue-derived extracellular vesicles in inter-organ crosstalk: Implications for metabolic regulation and adipose tissue function. Cell Rep 2025; 44:115732. [PMID: 40408250 DOI: 10.1016/j.celrep.2025.115732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 04/24/2025] [Accepted: 05/01/2025] [Indexed: 05/25/2025] Open
Abstract
Intercellular and inter-organ communication systems are vital for tissue homeostasis and disease development, utilizing soluble bioactive molecules for signaling. The field of extracellular vesicle (EV) biology has rapidly expanded in recent decades, highlighting EVs as effective bioactive nanovectors for cell-to-cell communication in various physiological and pathological contexts. Numerous studies indicate that adipocyte-derived EVs are crucial components of the adipose secretome, playing a key role in autocrine and paracrine interactions within adipose tissue, as well as in endocrine signaling. This review aims to present an updated perspective on EVs as mediators of communication between adipose tissue and other organs, while also examining their therapeutic potential in the light of recent advancements in EV biology research.
Collapse
Affiliation(s)
- Soazig Le Lay
- Nantes Université, CNRS, INSERM, l'institut du thorax, 44000 Nantes, France; Université Angers, SFR ICAT, 49000 Angers, France.
| | - Philipp E Scherer
- Touchstone Diabetes Center, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
2
|
Duan H, Siadat SH, Jasim SA, Bansal P, Kaur H, Qasim MT, Abosaoda MK, Aboqader Al-Aouadi RF, Suliman M, Ali Khiavi P. Therapeutic Potential of Exosomal miRNAs: New Insights and Future Directions. J Biochem Mol Toxicol 2025; 39:e70270. [PMID: 40272032 DOI: 10.1002/jbt.70270] [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: 12/24/2024] [Revised: 03/13/2025] [Accepted: 04/10/2025] [Indexed: 04/25/2025]
Abstract
Modern advancements in medicine include developing targeted drug delivery systems in the medical field, which are designed to unravel the potential of therapeutic products and overcome the barriers to the effectiveness of current approaches. Various nanopolymer carrier systems have been introduced in this regard, and the simple characteristics of extracellular vesicles have drawn special attention to their application as an effective drug delivery tool. Exosomes are very similar to transport vesicles and have a lipid-biomembrane covering an aqueous core. They also contain both hydrophilic and lipophilic substances and deliver their cargo to the desired targets. These properties enable exosomes to overcome some of the limitations of liposomes. Exosomes can easily diffuse into body fluids and remain in the bloodstream for a long time, crossing physiological barriers and entering cells. Exosomes, which contain a large volume of biomolecules, do not stimulate immune responses and do not accumulate in the liver or lungs instead of target tissues. Recent advancements in regenerative medicine have enabled scientists to utilize exosomes extracted from mesenchymal stem cells (MSCs), which possess significant regenerative abilities, for treating various diseases. The contents of these exosomes are crucial for both diagnosis and treatment, as they influence disease progression. Numerous in vitro studies have confirmed the safety, effectiveness, and therapeutic promise of exosomes in conditions such as cancer, neurodegenerative disorders, cardiovascular issues, and orthopedic ailments. This article explores the therapeutic potential of MSC-derived exosomes and outlines the essential procedures for their preparation.
Collapse
Affiliation(s)
- Haili Duan
- China Three Gorges University, Yichang City, China
| | | | - Saade Abdalkareem Jasim
- Medical Laboratory Techniques department, College of Health and medical technology, University of Al-maarif, Anbar, Iraq
| | - Pooja Bansal
- Department of Biotechnology and Genetics, Jain (Deemed-to-be) University, Bengaluru, India
- Department of Allied Healthcare and Sciences, Vivekananda Global University, Jaipur, India
| | - Harpreet Kaur
- School of Basic & Applied Sciences, Shobhit University, Gangoh, India
- Department of Health & Allied Sciences, Arka Jain University, Jamshedpur, India
| | - Maytham T Qasim
- Immunology and Physiology, College of Health and Medical Technology, Al-Ayen University, Iraq
| | - Munther Kadhim Abosaoda
- College of Pharmacy, The Islamic University, Najaf, Iraq
- College of Pharmacy, The Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
- College of Pharmacy, The Islamic University of Babylon, Al Diwaniyah, Iraq
| | | | - Muath Suliman
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Payam Ali Khiavi
- Medicine Faculty, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
3
|
Wen X, Hao Z, Yin H, Min J, Wang X, Sun S, Ruan G. Engineered Extracellular Vesicles as a New Class of Nanomedicine. CHEM & BIO ENGINEERING 2025; 2:3-22. [PMID: 39975802 PMCID: PMC11835263 DOI: 10.1021/cbe.4c00122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Revised: 10/19/2024] [Accepted: 10/20/2024] [Indexed: 02/21/2025]
Abstract
Extracellular vesicles (EVs) are secreted from biological cells and contain many molecules with diagnostic values or therapeutic functions. There has been great interest in academic and industrial communities to utilize EVs as tools for diagnosis or therapeutics. In addition, EVs can also serve as delivery vehicles for therapeutic molecules. An indicator of the enormous interest in EVs is the large number of review articles published on EVs, with the focus ranging from their biology to their applications. An emerging trend in EV research is to produce and utilize "engineered EVs", which are essentially the enhanced version of EVs. EV engineering can be conducted by cell culture condition control, genetic engineering, or chemical engineering. Given their nanometer-scale sizes and therapeutic potentials, engineered EVs are an emerging class of nanomedicines. So far, an overwhelming majority of the research on engineered EVs is preclinical studies; there are only a very small number of reported clinical trials. This Review focuses on engineered EVs, with a more specific focus being their applications in therapeutics. The various approaches to producing engineered EVs and their applications in various diseases are reviewed. Furthermore, in vivo imaging of EVs, the mechanistic understandings, and the clinical translation aspects are discussed. The discussion is primarily on preclinical studies while briefly mentioning the clinical trials. With continued interdisciplinary research efforts from biologists, pharmacists, physicians, bioengineers, and chemical engineers, engineered EVs could become a powerful solution for many major diseases such as neurological, immunological, and cardiovascular diseases.
Collapse
Affiliation(s)
- Xiaowei Wen
- Institute
of Analytical Chemistry and Instrument for Life Science, The Key Laboratory
of Biomedical Information Engineering of Ministry of Education, School
of Life Science and Technology, Xi’an
Jiaotong University, Xi’an, China 710049
- Wisdom
Lake Academy of Pharmacy, Xi’an Jiaotong-Liverpool
University, Suzhou, China 215123
- Jiangsu
Province Higher Education Key Laboratory of Cell Therapy Nanoformulation
(Construction), Xi’an Jiaotong-Liverpool
University, Suzhou, China 215123
- Xi’an
Jiaotong-Liverpool University & University of Liverpool Joint
Center of Pharmacology and Therapeutics, Suzhou, China 215123
| | - Zerun Hao
- Wisdom
Lake Academy of Pharmacy, Xi’an Jiaotong-Liverpool
University, Suzhou, China 215123
- Jiangsu
Province Higher Education Key Laboratory of Cell Therapy Nanoformulation
(Construction), Xi’an Jiaotong-Liverpool
University, Suzhou, China 215123
- Xi’an
Jiaotong-Liverpool University & University of Liverpool Joint
Center of Pharmacology and Therapeutics, Suzhou, China 215123
| | - Haofan Yin
- Wisdom
Lake Academy of Pharmacy, Xi’an Jiaotong-Liverpool
University, Suzhou, China 215123
- Jiangsu
Province Higher Education Key Laboratory of Cell Therapy Nanoformulation
(Construction), Xi’an Jiaotong-Liverpool
University, Suzhou, China 215123
- Xi’an
Jiaotong-Liverpool University & University of Liverpool Joint
Center of Pharmacology and Therapeutics, Suzhou, China 215123
| | - Jie Min
- Wisdom
Lake Academy of Pharmacy, Xi’an Jiaotong-Liverpool
University, Suzhou, China 215123
- Jiangsu
Province Higher Education Key Laboratory of Cell Therapy Nanoformulation
(Construction), Xi’an Jiaotong-Liverpool
University, Suzhou, China 215123
- Xi’an
Jiaotong-Liverpool University & University of Liverpool Joint
Center of Pharmacology and Therapeutics, Suzhou, China 215123
| | - Xueying Wang
- Wisdom
Lake Academy of Pharmacy, Xi’an Jiaotong-Liverpool
University, Suzhou, China 215123
- Jiangsu
Province Higher Education Key Laboratory of Cell Therapy Nanoformulation
(Construction), Xi’an Jiaotong-Liverpool
University, Suzhou, China 215123
- Xi’an
Jiaotong-Liverpool University & University of Liverpool Joint
Center of Pharmacology and Therapeutics, Suzhou, China 215123
| | - Sihan Sun
- Wisdom
Lake Academy of Pharmacy, Xi’an Jiaotong-Liverpool
University, Suzhou, China 215123
- Jiangsu
Province Higher Education Key Laboratory of Cell Therapy Nanoformulation
(Construction), Xi’an Jiaotong-Liverpool
University, Suzhou, China 215123
- Xi’an
Jiaotong-Liverpool University & University of Liverpool Joint
Center of Pharmacology and Therapeutics, Suzhou, China 215123
| | - Gang Ruan
- Wisdom
Lake Academy of Pharmacy, Xi’an Jiaotong-Liverpool
University, Suzhou, China 215123
- Jiangsu
Province Higher Education Key Laboratory of Cell Therapy Nanoformulation
(Construction), Xi’an Jiaotong-Liverpool
University, Suzhou, China 215123
- Xi’an
Jiaotong-Liverpool University & University of Liverpool Joint
Center of Pharmacology and Therapeutics, Suzhou, China 215123
| |
Collapse
|
4
|
Ahmad S, Christova T, Pye M, Narimatsu M, Song S, Wrana JL, Attisano L. Small Extracellular Vesicles Promote Axon Outgrowth by Engaging the Wnt-Planar Cell Polarity Pathway. Cells 2025; 14:56. [PMID: 39791757 PMCID: PMC11720052 DOI: 10.3390/cells14010056] [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: 10/20/2024] [Revised: 12/14/2024] [Accepted: 12/17/2024] [Indexed: 01/30/2025] Open
Abstract
In neurons, the acquisition of a polarized morphology is achieved upon the outgrowth of a single axon from one of several neurites. Small extracellular vesicles (sEVs), such as exosomes, from diverse sources are known to promote neurite outgrowth and thus may have therapeutic potential. However, the effect of fibroblast-derived exosomes on axon elongation in neurons of the central nervous system under growth-permissive conditions remains unclear. Here, we show that fibroblast-derived sEVs promote axon outgrowth and a polarized neuronal morphology in mouse primary embryonic cortical neurons. Mechanistically, we demonstrate that the sEV-induced increase in axon outgrowth requires endogenous Wnts and core PCP components including Prickle, Vangl, Frizzled, and Dishevelled. We demonstrate that sEVs are internalized by neurons, colocalize with Wnt7b, and induce relocalization of Vangl2 to the distal axon during axon outgrowth. In contrast, sEVs derived from neurons or astrocytes do not promote axon outgrowth, while sEVs from activated astrocytes inhibit elongation. Thus, our data reveal that fibroblast-derived sEVs promote axon elongation through the Wnt-PCP pathway in a manner that is dependent on endogenous Wnts.
Collapse
Affiliation(s)
- Samar Ahmad
- Department of Biochemistry, Donnelly Centre, University of Toronto, Toronto, ON M5S 3E1, Canada; (S.A.); (T.C.); (S.S.)
| | - Tania Christova
- Department of Biochemistry, Donnelly Centre, University of Toronto, Toronto, ON M5S 3E1, Canada; (S.A.); (T.C.); (S.S.)
| | - Melanie Pye
- Center for Systems Biology, Lunenfeld-Tanenbaum Research Institute, Mt. Sinai Hospital, Toronto, ON M5G 1X5, Canada; (M.P.); (M.N.); (J.L.W.)
| | - Masahiro Narimatsu
- Center for Systems Biology, Lunenfeld-Tanenbaum Research Institute, Mt. Sinai Hospital, Toronto, ON M5G 1X5, Canada; (M.P.); (M.N.); (J.L.W.)
| | - Siyuan Song
- Department of Biochemistry, Donnelly Centre, University of Toronto, Toronto, ON M5S 3E1, Canada; (S.A.); (T.C.); (S.S.)
| | - Jeffrey L. Wrana
- Center for Systems Biology, Lunenfeld-Tanenbaum Research Institute, Mt. Sinai Hospital, Toronto, ON M5G 1X5, Canada; (M.P.); (M.N.); (J.L.W.)
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5G 1X5, Canada
| | - Liliana Attisano
- Department of Biochemistry, Donnelly Centre, University of Toronto, Toronto, ON M5S 3E1, Canada; (S.A.); (T.C.); (S.S.)
| |
Collapse
|
5
|
Goldbloom-Helzner L, Bains H, Loll EG, Henson T, Mizenko RR, Kumar P, Tan C, Farmer DL, Carney RP, Wang A. Assessing the conjugation efficiency of surface-modified extracellular vesicles using single nanovesicle analysis technologies. NANOSCALE 2024; 16:20903-20916. [PMID: 39310954 PMCID: PMC11748815 DOI: 10.1039/d4nr01603c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2024]
Abstract
Extracellular vesicles (EVs) are cell-secreted nanoscale vesicles with important roles in cell-cell communication and drug delivery. Although EVs pose a promising alternative to cell-based therapy, targeted delivery in vivo is lacking. Their surface is often modified to endow them with active targeting molecules to enable specific cell uptake and tailor EV biodistribution. A dominant paradigm has been to evaluate the EV surface functionalization using bulk analysis assays, such as western blotting and bead-based flow cytometry. Yet, the heterogeneity of EVs is now recognized as a major bottleneck for their clinical translation. Here, we engineer the EV surface at the single-vesicle level. We applied orthogonal platforms with single vesicle resolution to determine and optimize the efficiency of conjugating the myelin-targeting aptamer LJM-3064 to single EVs (Apt-EVs). The aptamers were conjugated using either lipid insertion or covalent protein modification, followed by an assessment of single-EV integrity and stability. We observed unique aptamer conjugation to single EVs that depends on EV size. Our study underscores the importance of single vesicle analysis for engineering EVs and provides a novel single-EV-based framework for modifying EV surfaces.
Collapse
Affiliation(s)
- Leora Goldbloom-Helzner
- Center for Surgical Bioengineering, Department of Surgery, School of Medicine, University of California-Davis, Sacramento, CA, 95817, USA.
- Institute for Pediatric Regenerative Medicine, Shriners Children's, Sacramento, CA, 95817, USA
- Department of Biomedical Engineering, University of California-Davis, Davis, CA, 95616, USA
| | - Harjn Bains
- Department of Biomedical Engineering, University of California-Davis, Davis, CA, 95616, USA
| | - Emma G Loll
- Center for Surgical Bioengineering, Department of Surgery, School of Medicine, University of California-Davis, Sacramento, CA, 95817, USA.
- Institute for Pediatric Regenerative Medicine, Shriners Children's, Sacramento, CA, 95817, USA
| | - Tanner Henson
- Center for Surgical Bioengineering, Department of Surgery, School of Medicine, University of California-Davis, Sacramento, CA, 95817, USA.
- Department of Biomedical Engineering, University of California-Davis, Davis, CA, 95616, USA
| | - Rachel R Mizenko
- Department of Biomedical Engineering, University of California-Davis, Davis, CA, 95616, USA
| | - Priyadarsini Kumar
- Center for Surgical Bioengineering, Department of Surgery, School of Medicine, University of California-Davis, Sacramento, CA, 95817, USA.
- Institute for Pediatric Regenerative Medicine, Shriners Children's, Sacramento, CA, 95817, USA
| | - Cheemeng Tan
- Department of Biomedical Engineering, University of California-Davis, Davis, CA, 95616, USA
| | - Diana L Farmer
- Center for Surgical Bioengineering, Department of Surgery, School of Medicine, University of California-Davis, Sacramento, CA, 95817, USA.
- Institute for Pediatric Regenerative Medicine, Shriners Children's, Sacramento, CA, 95817, USA
| | - Randy P Carney
- Department of Biomedical Engineering, University of California-Davis, Davis, CA, 95616, USA
| | - Aijun Wang
- Center for Surgical Bioengineering, Department of Surgery, School of Medicine, University of California-Davis, Sacramento, CA, 95817, USA.
- Institute for Pediatric Regenerative Medicine, Shriners Children's, Sacramento, CA, 95817, USA
- Department of Biomedical Engineering, University of California-Davis, Davis, CA, 95616, USA
| |
Collapse
|
6
|
Elashry MI, Speer J, De Marco I, Klymiuk MC, Wenisch S, Arnhold S. Extracellular Vesicles: A Novel Diagnostic Tool and Potential Therapeutic Approach for Equine Osteoarthritis. Curr Issues Mol Biol 2024; 46:13078-13104. [PMID: 39590374 PMCID: PMC11593097 DOI: 10.3390/cimb46110780] [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: 10/13/2024] [Revised: 11/12/2024] [Accepted: 11/15/2024] [Indexed: 11/28/2024] Open
Abstract
Osteoarthritis (OA) is a chronic progressive degenerative joint disease that affects a significant portion of the equine population and humans worldwide. Current treatment options for equine OA are limited and incompletely curative. Horses provide an excellent large-animal model for studying human OA. Recent advances in the field of regenerative medicine have led to the exploration of extracellular vesicles (EVs)-cargoes of microRNA, proteins, lipids, and nucleic acids-to evaluate their diagnostic value in terms of disease progression and severity, as well as a potential cell-free therapeutic approach for equine OA. EVs transmit molecular signals that influence various biological processes, including the inflammatory response, apoptosis, proliferation, and cell communication. In the present review, we summarize recent advances in the isolation and identification of EVs, the use of their biologically active components as biomarkers, and the distribution of the gap junction protein connexin 43. Moreover, we highlight the role of mesenchymal stem cell-derived EVs as a potential therapeutic tool for equine musculoskeletal disorders. This review aims to provide a comprehensive overview of the current understanding of the pathogenesis, diagnosis, and treatment strategies for OA. In particular, the roles of EVs as biomarkers in synovial fluid, chondrocytes, and plasma for the early detection of equine OA are discussed.
Collapse
Affiliation(s)
- Mohamed I. Elashry
- Institute of Veterinary Anatomy, Histology and Embryology, Justus-Liebig-University of Giessen, 35392 Giessen, Germany; (J.S.); (M.C.K.); (S.A.)
| | - Julia Speer
- Institute of Veterinary Anatomy, Histology and Embryology, Justus-Liebig-University of Giessen, 35392 Giessen, Germany; (J.S.); (M.C.K.); (S.A.)
| | - Isabelle De Marco
- Clinic of Small Animals, c/o Institute of Veterinary Anatomy, Histology and Embryology, Justus-Liebig-University of Giessen, 35392 Giessen, Germany; (I.D.M.); (S.W.)
| | - Michele C. Klymiuk
- Institute of Veterinary Anatomy, Histology and Embryology, Justus-Liebig-University of Giessen, 35392 Giessen, Germany; (J.S.); (M.C.K.); (S.A.)
| | - Sabine Wenisch
- Clinic of Small Animals, c/o Institute of Veterinary Anatomy, Histology and Embryology, Justus-Liebig-University of Giessen, 35392 Giessen, Germany; (I.D.M.); (S.W.)
| | - Stefan Arnhold
- Institute of Veterinary Anatomy, Histology and Embryology, Justus-Liebig-University of Giessen, 35392 Giessen, Germany; (J.S.); (M.C.K.); (S.A.)
| |
Collapse
|
7
|
Li S, Li Y, Zhu K, He W, Guo X, Wang T, Gong S, Zhu Z. Exosomes from mesenchymal stem cells: Potential applications in wound healing. Life Sci 2024; 357:123066. [PMID: 39306326 DOI: 10.1016/j.lfs.2024.123066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 07/08/2024] [Accepted: 09/16/2024] [Indexed: 09/26/2024]
Abstract
Wound healing is a continuous and complex process regulated by multiple factors, which has become an intractable clinical burden. Mesenchymal stem cell-derived exosomes (MSC-exos) possess low immunogenicity, easy preservation, and potent bioactivity, which is a mirror to their parental cells MSC-exos are important tools for regulating the biological behaviors of wound healing-associated cells, including fibroblasts, keratinocytes, immune cells, and endothelial cells. MSC-exos accelerate the wound healing process at cellular and animal levels by modulating inflammatory responses, promoting collagen deposition and vascularization. MSC-exos accelerate wound healing at the cellular and animal levels by modulating inflammatory responses and promoting collagen deposition and vascularization. This review summarizes the roles and mechanisms of MSC-exos originating from various sources in promoting the healing efficacy of general wounds, diabetic wounds, burn wounds, and healing-related scars. It also discusses the limitations and perspectives of MSC-exos in wound healing, in terms of exosome acquisition, mechanistic complexity, and exosome potentiation modalities. A deeper understanding of the properties and functions of MSC-exos is beneficial to advance the therapeutic approaches for achieving optimal wound healing.
Collapse
Affiliation(s)
- Sicheng Li
- Department of Plastic Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei Province, China
| | - Yichuan Li
- Department of Dermatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Keyu Zhu
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Wenlin He
- Department of Plastic Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei Province, China
| | - Xingjun Guo
- Department of Biliary-Pancreatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Ting Wang
- Department of Medical Ultrasound, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China.
| | - Song Gong
- Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Zhanyong Zhu
- Department of Plastic Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei Province, China.
| |
Collapse
|
8
|
Zhou R, Huang R, Xu Y, Zhang D, Gu L, Su Y, Chen X, Shi W, Sun J, Gu P, Ni N, Bi X. Exosomes derived from mucoperiosteum Krt14 +Ctsk + cells promote bone regeneration by coupling enhanced osteogenesis and angiogenesis. Biomater Sci 2024; 12:5753-5765. [PMID: 39392433 DOI: 10.1039/d4bm00673a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Repair of large bone defects is a sophisticated physiological process involving the meticulous orchestration of cell activation, proliferation, and differentiation. Cellular interactions between different cell types are paramount for successful bone regeneration, making it a challenging yet fascinating area of research and clinical practice. With increasing evidence underscoring the essential role of exosomes in facilitating intercellular and cell-microenvironment communication, they have emerged as an encouraging therapeutic strategy to promote bone repair due to their non-immunogenicity, diverse sources, and potent bioactivity. In this study, we characterized a distinctive population of Krt14+Ctsk+ cells from the orbital mucoperiosteum. In vitro experiments confirmed that exosomes from Krt14+Ctsk+ cells dramatically boosted the capacities of human umbilical vein endothelial cells (HUVECs) to proliferate, migrate, and induce angiogenesis. Additionally, the exosomes notably elevated the expression of osteogenic markers, thereby indicating their potential to augment osteogenic capabilities. Furthermore, in vivo experiments utilizing a rat calvarial defect model verified that exosome-loaded sodium alginate (SA) hydrogels accelerated local vascularized bone regeneration within the defective regions. Collectively, these findings suggest that exosomes secreted by Krt14+Ctsk+ cells offer an innovative method to accelerate bone repair via coupling enhanced osteogenesis and angiogenesis, highlighting the therapeutic potential in bone repair.
Collapse
Affiliation(s)
- Rong Zhou
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, P.R. China.
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200011, P.R. China
- Center for Basic Medical Research and Innovation in Visual System Diseases, Ministry of Education, China
| | - Rui Huang
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, P.R. China.
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200011, P.R. China
- Center for Basic Medical Research and Innovation in Visual System Diseases, Ministry of Education, China
| | - Yue Xu
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, P.R. China.
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200011, P.R. China
- Center for Basic Medical Research and Innovation in Visual System Diseases, Ministry of Education, China
| | - Dandan Zhang
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, P.R. China.
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200011, P.R. China
- Center for Basic Medical Research and Innovation in Visual System Diseases, Ministry of Education, China
| | - Li Gu
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, P.R. China.
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200011, P.R. China
- Center for Basic Medical Research and Innovation in Visual System Diseases, Ministry of Education, China
| | - Yun Su
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, P.R. China.
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200011, P.R. China
- Center for Basic Medical Research and Innovation in Visual System Diseases, Ministry of Education, China
| | - Xirui Chen
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, P.R. China.
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200011, P.R. China
- Center for Basic Medical Research and Innovation in Visual System Diseases, Ministry of Education, China
| | - Wodong Shi
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, P.R. China.
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200011, P.R. China
- Center for Basic Medical Research and Innovation in Visual System Diseases, Ministry of Education, China
| | - Jing Sun
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, P.R. China.
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200011, P.R. China
- Center for Basic Medical Research and Innovation in Visual System Diseases, Ministry of Education, China
| | - Ping Gu
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, P.R. China.
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200011, P.R. China
- Center for Basic Medical Research and Innovation in Visual System Diseases, Ministry of Education, China
| | - Ni Ni
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, P.R. China.
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200011, P.R. China
- Center for Basic Medical Research and Innovation in Visual System Diseases, Ministry of Education, China
| | - Xiaoping Bi
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, P.R. China.
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200011, P.R. China
- Center for Basic Medical Research and Innovation in Visual System Diseases, Ministry of Education, China
| |
Collapse
|
9
|
Zhang Q, Liu J, Wang W, Lin W, Ahmed W, Duan W, Huang S, Zhu Z, Chen L. The role of exosomes derived from stem cells in nerve regeneration: A contribution to neurological repair. Exp Neurol 2024; 380:114882. [PMID: 39002923 DOI: 10.1016/j.expneurol.2024.114882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 06/27/2024] [Accepted: 07/08/2024] [Indexed: 07/15/2024]
Abstract
Stem cell-derived exosomes have gained attention in regenerative medicine for their role in encouraging nerve regeneration and potential use in treating neurological diseases. These nanosized extracellular vesicles act as carriers of bioactive molecules, facilitating intercellular communication and enhancing the regenerative process in neural tissues. This comprehensive study explores the methods by which exosomes produced from various stem cells contribute to nerve healing, with a particular emphasis on their role in angiogenesis, inflammation, and cellular signaling pathways. By examining cutting-edge developments and exploring the potential of exosomes in delivering disease-specific miRNAs and proteins, we highlight their versatility in tailoring personalized therapeutic strategies. The findings presented here highlight the potential of stem cell-produced exosomes for use in neurological diseases therapy, establishing the door for future research into exosome-based neurotherapies.
Collapse
Affiliation(s)
- Qiankun Zhang
- Department of Neurosurgery, Southern Medical University Hospital of Integrated Traditional Chinese and Western Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Jiale Liu
- Department of Neurosurgery, Southern Medical University Hospital of Integrated Traditional Chinese and Western Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Wei Wang
- Department of Neurosurgery, Southern Medical University Hospital of Integrated Traditional Chinese and Western Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Wentong Lin
- Department of Orthopaedics, Chaozhou Hospital of Traditional Chinese Medicine, Chaozhou, China
| | - Waqas Ahmed
- School of Medicine, Southeast University, Nanjing, Jiangsu, China
| | - Wenjie Duan
- Department of Neurosurgery, Southern Medical University Hospital of Integrated Traditional Chinese and Western Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Songze Huang
- Department of Neurosurgery, Southern Medical University Hospital of Integrated Traditional Chinese and Western Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Zhihan Zhu
- Department of Neurosurgery, Southern Medical University Hospital of Integrated Traditional Chinese and Western Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Lukui Chen
- Department of Neurosurgery, Southern Medical University Hospital of Integrated Traditional Chinese and Western Medicine, Southern Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
10
|
Mizenko RR, Feaver M, Bozkurt BT, Lowe N, Nguyen B, Huang K, Wang A, Carney RP. A critical systematic review of extracellular vesicle clinical trials. J Extracell Vesicles 2024; 13:e12510. [PMID: 39330928 PMCID: PMC11428870 DOI: 10.1002/jev2.12510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 08/06/2024] [Accepted: 09/06/2024] [Indexed: 09/28/2024] Open
Abstract
This systematic review examines the landscape of extracellular vesicle (EV)-related clinical trials to elucidate the field's trends in clinical applications and EV-related methodologies, with an additional focus on the acknowledgement of EV subpopulations. By analysing data from public reporting repositories, we catalogued 471 EV-related clinical trials to date, with indications for over 200 diseases. Diagnostics and companion diagnostics represented the bulk of EV-related clinical trials with cancer being the most frequent application. EV-related therapeutics trials mainly utilized mesenchymal stromal cell (MSC) EVs and were most frequently used for treatment of respiratory illnesses. Ultracentrifugation and RNA-sequencing were the most common isolation and characterization techniques; however, methodology for each was not frequently reported in study records. Most of the reported characterization relied on bulk characterization of EV isolates, with only 11% utilizing EV subpopulations in their experimental design. While this may be connected to a lack of available techniques suitable for clinical implementation, it also highlights the opportunity for use of EV subpopulations to improve translational efforts. As academic research identifies more chemically distinct subpopulations and technologies for their enrichment, we forecast to more refined EV trials in the near future. This review emphasizes the need for meticulous methodological reporting and consideration of EV subpopulations to enhance the translational success of EV-based interventions, pointing towards a paradigm shift in personalized medicine.
Collapse
Affiliation(s)
- Rachel R. Mizenko
- Department of Biomedical EngineeringUniversity of CaliforniaDavisCaliforniaUSA
| | - Madison Feaver
- Department of Biomedical EngineeringUniversity of CaliforniaDavisCaliforniaUSA
| | - Batuhan T. Bozkurt
- Department of Biomedical EngineeringUniversity of CaliforniaDavisCaliforniaUSA
| | - Neona Lowe
- Department of Biomedical EngineeringUniversity of CaliforniaDavisCaliforniaUSA
| | - Bryan Nguyen
- Department of Biomedical EngineeringUniversity of CaliforniaDavisCaliforniaUSA
| | - Kuan‐Wei Huang
- Department of Biomedical EngineeringUniversity of CaliforniaDavisCaliforniaUSA
| | - Aijun Wang
- Department of Biomedical EngineeringUniversity of CaliforniaDavisCaliforniaUSA
- Department of SurgeryUniversity of CaliforniaDavisCaliforniaUSA
| | - Randy P. Carney
- Department of Biomedical EngineeringUniversity of CaliforniaDavisCaliforniaUSA
| |
Collapse
|
11
|
Pedersen C, Chen VT, Herbst P, Zhang R, Elfert A, Krishan A, Azar DT, Chang JH, Hu WY, Kremsmayer TP, Jalilian E, Djalilian AR, Guaiquil VH, Rosenblatt MI. Target specification and therapeutic potential of extracellular vesicles for regulating corneal angiogenesis, lymphangiogenesis, and nerve repair. Ocul Surf 2024; 34:459-476. [PMID: 39426677 PMCID: PMC11921040 DOI: 10.1016/j.jtos.2024.10.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 09/16/2024] [Accepted: 10/15/2024] [Indexed: 10/21/2024]
Abstract
Extracellular vesicles, including exosomes, are small extracellular vesicles that range in size from 30 nm to 10 μm in diameter and have specific membrane markers. They are naturally secreted and are present in various bodily fluids, including blood, urine, and saliva, and through the variety of their internal cargo, they contribute to both normal physiological and pathological processes. These processes include immune modulation, neuronal synapse formation, cell differentiation, cancer metastasis, angiogenesis, lymphangiogenesis, progression of infectious disease, and neurodegenerative disorders like Alzheimer's and Parkinson's disease. In recent years, interest has grown in the use of exosomes as a potential drug delivery system for various diseases and injuries. Importantly, exosomes originating from a patient's own cells exhibit minimal immunogenicity and possess remarkable stability along with inherent and adjustable targeting capabilities. This review explores the roles of exosomes in angiogenesis, lymphangiogenesis, and nerve repair with a specific emphasis on these processes within the cornea. Furthermore, it examines exosomes derived from specific cell types, discusses the advantages of exosome-based therapies in modulating these processes, and presents some of the most established methods for exosome isolation. Exosome-based treatments are emerging as potential minimally invasive and non-immunogenic therapies that modulate corneal angiogenesis and lymphangiogenesis, as well as enhance and accelerate endogenous corneal nerve repair.
Collapse
Affiliation(s)
- Cameron Pedersen
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Victoria T Chen
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Paula Herbst
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Runze Zhang
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Amr Elfert
- University of Illinois Cancer Center, Chicago, IL, USA
| | - Abhi Krishan
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Dimitri T Azar
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Jin-Hong Chang
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA; Jesse Brown Veterans Affairs Medical Center, Chicago, IL, USA.
| | - Wen-Yang Hu
- Department of Urology, University of Illinois at Chicago, Chicago, IL, USA
| | - Tobias P Kremsmayer
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Elmira Jalilian
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA; Richard and Loan Hill Department of Bioengineering, University of Illinois at Chicago, Chicago, IL, 60607, USA
| | - Ali R Djalilian
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Victor H Guaiquil
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Mark I Rosenblatt
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA.
| |
Collapse
|
12
|
Putthanbut N, Lee JY, Borlongan CV. Extracellular vesicle therapy in neurological disorders. J Biomed Sci 2024; 31:85. [PMID: 39183263 PMCID: PMC11346291 DOI: 10.1186/s12929-024-01075-w] [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: 06/17/2024] [Accepted: 08/16/2024] [Indexed: 08/27/2024] Open
Abstract
Extracellular vesicles (EVs) are vital for cell-to-cell communication, transferring proteins, lipids, and nucleic acids in various physiological and pathological processes. They play crucial roles in immune modulation and tissue regeneration but are also involved in pathogenic conditions like inflammation and degenerative disorders. EVs have heterogeneous populations and cargo, with numerous subpopulations currently under investigations. EV therapy shows promise in stimulating tissue repair and serving as a drug delivery vehicle, offering advantages over cell therapy, such as ease of engineering and minimal risk of tumorigenesis. However, challenges remain, including inconsistent nomenclature, complex characterization, and underdeveloped large-scale production protocols. This review highlights the recent advances and significance of EVs heterogeneity, emphasizing the need for a better understanding of their roles in disease pathologies to develop tailored EV therapies for clinical applications in neurological disorders.
Collapse
Affiliation(s)
- Napasiri Putthanbut
- Department of Neurosurgery, Center of Aging and Brain Repair, University of South Florida, Tampa, USA
- Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Salaya, Thailand
| | - Jea Young Lee
- Department of Neurosurgery, Center of Aging and Brain Repair, University of South Florida, Tampa, USA
| | - Cesario V Borlongan
- Department of Neurosurgery, Center of Aging and Brain Repair, University of South Florida, Tampa, USA.
| |
Collapse
|
13
|
Li Q, Zhang F, Fu X, Han N. Therapeutic Potential of Mesenchymal Stem Cell-Derived Exosomes as Nanomedicine for Peripheral Nerve Injury. Int J Mol Sci 2024; 25:7882. [PMID: 39063125 PMCID: PMC11277195 DOI: 10.3390/ijms25147882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Revised: 07/02/2024] [Accepted: 07/07/2024] [Indexed: 07/28/2024] Open
Abstract
Peripheral nerve injury (PNI) is a complex and protracted process, and existing therapeutic approaches struggle to achieve effective nerve regeneration. Recent studies have shown that mesenchymal stem cells (MSCs) may be a pivotal choice for treating peripheral nerve injury. MSCs possess robust paracrine capabilities, and exosomes, as the primary secretome of MSCs, are considered crucial regulatory mediators involved in peripheral nerve regeneration. Exosomes, as nanocarriers, can transport various endogenous or exogenous bioactive substances to recipient cells, thereby promoting vascular and axonal regeneration while suppressing inflammation and pain. In this review, we summarize the mechanistic roles of exosomes derived from MSCs in peripheral nerve regeneration, discuss the engineering strategies for MSC-derived exosomes to improve therapeutic potential, and explore the combined effects of MSC-derived exosomes with biomaterials (nerve conduits, hydrogels) in peripheral nerve regeneration.
Collapse
Affiliation(s)
- Qicheng Li
- Department of Trauma and Orthopedics, Peking University People’s Hospital, Beijing 100044, China; (Q.L.); (F.Z.); (X.F.)
- Key Laboratory of Trauma and Neural Regeneration, Peking University, Beijing 100044, China
| | - Fengshi Zhang
- Department of Trauma and Orthopedics, Peking University People’s Hospital, Beijing 100044, China; (Q.L.); (F.Z.); (X.F.)
- Key Laboratory of Trauma and Neural Regeneration, Peking University, Beijing 100044, China
| | - Xiaoyang Fu
- Department of Trauma and Orthopedics, Peking University People’s Hospital, Beijing 100044, China; (Q.L.); (F.Z.); (X.F.)
- Key Laboratory of Trauma and Neural Regeneration, Peking University, Beijing 100044, China
| | - Na Han
- Department of Trauma and Orthopedics, Peking University People’s Hospital, Beijing 100044, China; (Q.L.); (F.Z.); (X.F.)
- Key Laboratory of Trauma and Neural Regeneration, Peking University, Beijing 100044, China
- National Center for Trauma Medicine, Beijing 100044, China
| |
Collapse
|
14
|
Chen Y, Zhang Z, Li Z, Wu W, Lan S, Yan T, Mei K, Qiao Z, Wang C, Bai C, Li Z, Wu S, Wang J, Zhang Q. Dynamic nanomechanical characterization of cells in exosome therapy. MICROSYSTEMS & NANOENGINEERING 2024; 10:97. [PMID: 39015940 PMCID: PMC11251037 DOI: 10.1038/s41378-024-00735-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 03/20/2024] [Accepted: 05/23/2024] [Indexed: 07/18/2024]
Abstract
Exosomes derived from mesenchymal stem cells (MSCs) have been confirmed to enhance cell proliferation and improve tissue repair. Exosomes release their contents into the cytoplasmic solution of the recipient cell to mediate cell expression, which is the main pathway through which exosomes exert therapeutic effects. The corresponding process of exosome internalization mainly occurs in the early stage of treatment. However, the therapeutic effect of exosomes in the early stage remains to be further studied. We report that the three-dimensional cell traction force can intuitively reflect the ability of exosomes to enhance the cytoskeleton and cell contractility of recipient cells, serving as an effective method to characterize the therapeutic effect of exosomes. Compared with traditional biochemical methods, we can visualize the early therapeutic effect of exosomes in real time without damage by quantifying the cell traction force. Through quantitative analysis of traction forces, we found that endometrial stromal cells exhibit short-term cell roundness accompanied by greater traction force during the early stage of exosome therapy. Further experiments revealed that exosomes enhance the traction force and cytoskeleton by regulating the Rac1/RhoA signaling pathway, thereby promoting cell proliferation. This work provides an effective method for rapidly quantifying the therapeutic effects of exosomes and studying the underlying mechanisms involved.
Collapse
Affiliation(s)
- Ye Chen
- CAS Key Laboratory of Mechanical Behavior and Design of Material, Department of Modern Mechanics, University of Science and Technology of China, Hefei, Anhui 230027 China
| | - Zihan Zhang
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022 China
- Anhui Province Key Laboratory of Reproductive Health and Genetics, Anhui Medical University, Hefei, 230022 China
| | - Ziwei Li
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022 China
- Anhui Province Key Laboratory of Reproductive Health and Genetics, Anhui Medical University, Hefei, 230022 China
| | - Wenjie Wu
- CAS Key Laboratory of Mechanical Behavior and Design of Material, Department of Modern Mechanics, University of Science and Technology of China, Hefei, Anhui 230027 China
| | - Shihai Lan
- CAS Key Laboratory of Mechanical Behavior and Design of Material, Department of Modern Mechanics, University of Science and Technology of China, Hefei, Anhui 230027 China
| | - Tianhao Yan
- Department of Cell Biology and Genetics, College of Basic Medical Sciences, Jilin University, Changchun, 130021 China
| | - Kainan Mei
- CAS Key Laboratory of Mechanical Behavior and Design of Material, Department of Modern Mechanics, University of Science and Technology of China, Hefei, Anhui 230027 China
| | - Zihan Qiao
- CAS Key Laboratory of Mechanical Behavior and Design of Material, Department of Modern Mechanics, University of Science and Technology of China, Hefei, Anhui 230027 China
| | - Chen Wang
- CAS Key Laboratory of Mechanical Behavior and Design of Material, Department of Modern Mechanics, University of Science and Technology of China, Hefei, Anhui 230027 China
| | - Chuanbiao Bai
- CAS Key Laboratory of Mechanical Behavior and Design of Material, Department of Modern Mechanics, University of Science and Technology of China, Hefei, Anhui 230027 China
| | - Ziyan Li
- CAS Key Laboratory of Mechanical Behavior and Design of Material, Department of Modern Mechanics, University of Science and Technology of China, Hefei, Anhui 230027 China
| | - Shangquan Wu
- CAS Key Laboratory of Mechanical Behavior and Design of Material, Department of Modern Mechanics, University of Science and Technology of China, Hefei, Anhui 230027 China
- State Key Laboratory of Nonlinear Mechanics, Institute of Mechanics, Chinese Academy of Science, 15 Beisihuan West Road, Beijing, 100190 China
| | - Jianye Wang
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022 China
- Anhui Province Key Laboratory of Reproductive Health and Genetics, Anhui Medical University, Hefei, 230022 China
| | - Qingchuan Zhang
- CAS Key Laboratory of Mechanical Behavior and Design of Material, Department of Modern Mechanics, University of Science and Technology of China, Hefei, Anhui 230027 China
| |
Collapse
|
15
|
Wang K, Yang Z, Zhang B, Gong S, Wu Y. Adipose-Derived Stem Cell Exosomes Facilitate Diabetic Wound Healing: Mechanisms and Potential Applications. Int J Nanomedicine 2024; 19:6015-6033. [PMID: 38911504 PMCID: PMC11192296 DOI: 10.2147/ijn.s466034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 06/08/2024] [Indexed: 06/25/2024] Open
Abstract
Wound healing in diabetic patients is frequently hampered. Adipose-derived stem cell exosomes (ADSC-eoxs), serving as a crucial mode of intercellular communication, exhibit promising therapeutic roles in facilitating wound healing. This review aims to comprehensively outline the molecular mechanisms through which ADSC-eoxs enhance diabetic wound healing. We emphasize the biologically active molecules released by these exosomes and their involvement in signaling pathways associated with inflammation modulation, cellular proliferation, vascular neogenesis, and other pertinent processes. Additionally, the clinical application prospects of the reported ADSC-eoxs are also deliberated. A thorough understanding of these molecular mechanisms and potential applications is anticipated to furnish a theoretical groundwork for combating diabetic wound healing.
Collapse
Affiliation(s)
- Kang Wang
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Zihui Yang
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Boyu Zhang
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Song Gong
- Division of Endocrinology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Yiping Wu
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| |
Collapse
|
16
|
Wang LJ, Tsai CC, Chao HR, Lee SY, Chen CC, Li SC. MicroRNAs in Umbilical Cord Blood and Development in Full-Term Newborns: A Prospective Study. Biomark Insights 2024; 19:11772719241258017. [PMID: 38863527 PMCID: PMC11165956 DOI: 10.1177/11772719241258017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 05/10/2024] [Indexed: 06/13/2024] Open
Abstract
Background Exploring the epigenetic regulations, such as microRNA, in newborns holds significant promise for enhancing our ability to address and potentially prevent early-life developmental delays. Objectives Hence, this research seeks to investigate if the expression of miRNA in the umbilical cord blood of infants can forecast their developmental outcomes as they grow older. Design and method We enrolled 143 full-term newborns, delivered either via cesarean section (CS) or through natural spontaneous delivery (NSD). We then analyzed the profiles of specific miRNAs (miR-486-5p, miR-126-5p, miR-140-3p, miR-151a-3p, miR-142-5p, and miR-30e-5p) in the umbilical cord blood of these infants. Subsequently, we performed follow-up assessments using Bayley-III scores when the cohort reached 1 year of age. Furthermore, we conducted pathway-enrichment analyses on the target genes associated with these examined miRNAs. Results When comparing newborns delivered via cesarean section (CS) to those born via natural spontaneous delivery (NSD), we observed notable differences. Specifically, newborns through NSD displayed significantly higher ΔCt values for miR-486-5p, alongside lower ΔCt values for miR-126-5p and miR-151a-3p in their cord blood. At 1 year of age, cognitive development was significantly linked to the ΔCt values of miR-140-3p and miR-142-5p, while language development showed a significant association with the ΔCt values of miR-140-3p. Moreover, our pathway enrichment analyses revealed that the target genes of these miRNAs were consistently involved in the pathways related to neurons, such as axon guidance and the neurotrophin signaling pathway. Conclusion In summary, this study represents a pioneering effort in elucidating the potential connections between miRNA levels in cord blood and the health indicators and neurodevelopment of newborns at 1 year of age. Our findings underscore the significance of miRNA levels at birth in influencing mechanisms related to neurodevelopment.
Collapse
Affiliation(s)
- Liang-Jen Wang
- Department of Child and Adolescent Psychiatry, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Ching-Chang Tsai
- Department of Obstetrics and Gynecology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - How-Ran Chao
- Department of Environmental Science and Engineering, College of Engineering, National Pingtung University of Science and Technology, Pingtung County, Taiwan
| | - Sheng-Yu Lee
- Department of Psychiatry, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
- Department of Psychiatry, College of Medicine, Graduate Institute of Medicine, School of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chih-Cheng Chen
- Section of Neonatology, Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
- Department of Early Childhood Care and Education, Cheng-Shiu University, Kaohsiung, Taiwan
- School of Medicine, College of Medicine, National Sun Yat-Sen University, Kaohsiung, Taiwan
| | - Sung-Chou Li
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
- Department of Dental Technology, Shu-Zen Junior College of Medicine and Management, Kaohsiung, Taiwan
| |
Collapse
|
17
|
Gul R, Bashir H, Sarfraz M, Shaikh AJ, Bin Jardan YA, Hussain Z, Bin Asad MHH, Gulzar F, Guan B, Nazir I, Amirzada MI. Human plasma derived exosomes: Impact of active and passive drug loading approaches on drug delivery. Saudi Pharm J 2024; 32:102096. [PMID: 38757071 PMCID: PMC11097067 DOI: 10.1016/j.jsps.2024.102096] [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: 11/23/2023] [Accepted: 05/05/2024] [Indexed: 05/18/2024] Open
Abstract
The aim of the current study was to explore the potential of human plasma-derived exosomes as versatile carriers for drug delivery by employing various active and passive loading methods. Exosomes were isolated from human plasma using differential centrifugation and ultrafiltration method. Drug loading was achieved by employing sonication and freeze thaw methods, facilitating effective drug encapsulation within exosomes for delivery. Each approach was examined for its effectiveness, loading efficiency and ability to preserve membrane stability. Methotrexate (MTX), a weak acid model drug was loaded at a concentration of 2.2 µM to exosomes underwent characterization using various techniques such as particle size analysis, transmission electron microscopy and drug loading capacity. Human plasma derived exosomes showed a mean size of 162.15 ± 28.21 nm and zeta potential of -30.6 ± 0.71 mV. These exosomes were successfully loaded with MTX demonstrated a better drug encapsulation of 64.538 ± 1.54 % by freeze thaw method in comparison 55.515 ± 1.907 % by sonication. In-vitro drug release displayed 60 % loaded drug released within 72 h by freeze thaw method that was significantly different from that by sonication method i.e., 99 % within 72 h (p value 0.0045). Moreover, cell viability of exosomes loaded by freeze thaw method was significantly higher than that by sonication method (p value 0.0091) suggested that there was membrane disruption by sonication method. In conclusion, this study offers valuable insights into the potential of human plasma-derived exosomes loaded by freeze thaw method suggest as a promising carrier for improved drug loading and maintenance of exosomal membrane integrity.
Collapse
Affiliation(s)
- Rabia Gul
- Department of Pharmacy, COMSATS University Islamabad, Abbottabad Campus, 22060 Abbottabad, Pakistan
| | - Hamid Bashir
- Centre for Applied Molecular Biology, University of the Punjab, 54000 Lahore, Pakistan
| | - Muhammad Sarfraz
- College of Pharmacy, Al-Ain University, 64141 Al-Ain, United Arab Emirates
| | - Ahson Jabbar Shaikh
- Department of Chemistry, COMSATS University Islamabad, Abbottabad Campus, 22060 Abbottabad, Pakistan
| | - Yousef A. Bin Jardan
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Zahid Hussain
- Department of Pharmaceutics and Pharmaceutical Technology, College of Pharmacy, University of Sharjah, 27272 Sharjah, United Arab Emirates
| | | | - Faisal Gulzar
- Department of Pharmacy, The University of Chenab, 50700 Gujrat, Pakistan
| | - Bo Guan
- School of Food Science and Technology, Shihezi University, 832000 Shihezi, Xinjiang, China
| | - Imran Nazir
- Department of Pharmacy, COMSATS University Islamabad, Lahore Campus, 54000 Lahore, Pakistan
| | - Muhammad Imran Amirzada
- Department of Pharmacy, COMSATS University Islamabad, Abbottabad Campus, 22060 Abbottabad, Pakistan
| |
Collapse
|
18
|
Moghassemi S, Dadashzadeh A, Sousa MJ, Vlieghe H, Yang J, León-Félix CM, Amorim CA. Extracellular vesicles in nanomedicine and regenerative medicine: A review over the last decade. Bioact Mater 2024; 36:126-156. [PMID: 38450204 PMCID: PMC10915394 DOI: 10.1016/j.bioactmat.2024.02.021] [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: 12/01/2023] [Revised: 02/15/2024] [Accepted: 02/19/2024] [Indexed: 03/08/2024] Open
Abstract
Small extracellular vesicles (sEVs) are known to be secreted by a vast majority of cells. These sEVs, specifically exosomes, induce specific cell-to-cell interactions and can activate signaling pathways in recipient cells through fusion or interaction. These nanovesicles possess several desirable properties, making them ideal for regenerative medicine and nanomedicine applications. These properties include exceptional stability, biocompatibility, wide biodistribution, and minimal immunogenicity. However, the practical utilization of sEVs, particularly in clinical settings and at a large scale, is hindered by the expensive procedures required for their isolation, limited circulation lifetime, and suboptimal targeting capacity. Despite these challenges, sEVs have demonstrated a remarkable ability to accommodate various cargoes and have found extensive applications in the biomedical sciences. To overcome the limitations of sEVs and broaden their potential applications, researchers should strive to deepen their understanding of current isolation, loading, and characterization techniques. Additionally, acquiring fundamental knowledge about sEVs origins and employing state-of-the-art methodologies in nanomedicine and regenerative medicine can expand the sEVs research scope. This review provides a comprehensive overview of state-of-the-art exosome-based strategies in diverse nanomedicine domains, encompassing cancer therapy, immunotherapy, and biomarker applications. Furthermore, we emphasize the immense potential of exosomes in regenerative medicine.
Collapse
Affiliation(s)
- Saeid Moghassemi
- Pôle de Recherche en Physiopathologie de La Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Arezoo Dadashzadeh
- Pôle de Recherche en Physiopathologie de La Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Maria João Sousa
- Pôle de Recherche en Physiopathologie de La Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Hanne Vlieghe
- Pôle de Recherche en Physiopathologie de La Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Jie Yang
- Pôle de Recherche en Physiopathologie de La Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Cecibel María León-Félix
- Pôle de Recherche en Physiopathologie de La Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Christiani A. Amorim
- Pôle de Recherche en Physiopathologie de La Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| |
Collapse
|
19
|
Wang Y, Liu X, Wang B, Sun H, Ren Y, Zhang H. Compounding engineered mesenchymal stem cell-derived exosomes: A potential rescue strategy for retinal degeneration. Biomed Pharmacother 2024; 173:116424. [PMID: 38471273 DOI: 10.1016/j.biopha.2024.116424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Revised: 02/28/2024] [Accepted: 03/08/2024] [Indexed: 03/14/2024] Open
Abstract
The prevalence of retinal degenerative diseases, including age-related macular degeneration and retinitis pigmentosa, has been increasing globally and is linked to the aging population and improved life expectancy. These diseases are characterized by chronic, progressive neuronal damage or depletion of the photoreceptor cells in the retina, and limited effective treatment options are currently available. Mesenchymal stem cell-derived exosomes (MSC-EXOs) containing cytokines, growth factors, lipids, mRNA, and miRNA, which act as mediators of intercellular communication transferring bioactive molecules to recipient cells, offer an appealing, non-cellular nanotherapeutic approach for retinal degenerative diseases. However, treatment specificity is compromised due to their high heterogeneity in size, content, functional effects, and parental cellular source. To improve this, engineered MSC-EXOs with increased drug-loading capacity, targeting ability, and resistance to bodily degradation and elimination have been developed. This review summarizes the recent advances in miRNAs of MSC-EXOs as a treatment for retinal degeneration, discussing the strategies and methods for engineering therapeutic MSC-EXOs. Notably, to address the single functional role of engineered MSC-EXOs, we propose a novel concept called "Compound Engineered MSC-EXOs (Co-E-MSC-EXOs)" along with its derived potential therapeutic approaches. The advantages and challenges of employing Co-E-MSC-EXOs for retinal degeneration in clinical applications, as well as the strategies and issues related to them, are also highlighted.
Collapse
Affiliation(s)
- Yao Wang
- Shaanxi Provincial Clinical Research Center for Ophthalmology Diseases, the First Affiliated Hospital of Northwest University, Xi'an No.1 hospital, Xi'an, Shaanxi, China; Shaanxi Key Laboratory of Ophthalmology, Shaanxi Institute of Ophthalmology, Xi'an, Shaanxi 710002, China.
| | - Xianning Liu
- Shaanxi Provincial Clinical Research Center for Ophthalmology Diseases, the First Affiliated Hospital of Northwest University, Xi'an No.1 hospital, Xi'an, Shaanxi, China; Shaanxi Key Laboratory of Ophthalmology, Shaanxi Institute of Ophthalmology, Xi'an, Shaanxi 710002, China
| | - Bei Wang
- The College of Life Sciences, Northwest University, Xi'an, Shaanxi 710069, China
| | - Hanhan Sun
- The College of Life Sciences, Northwest University, Xi'an, Shaanxi 710069, China
| | - Yiqian Ren
- Shaanxi Provincial Clinical Research Center for Ophthalmology Diseases, the First Affiliated Hospital of Northwest University, Xi'an No.1 hospital, Xi'an, Shaanxi, China; Shaanxi Key Laboratory of Ophthalmology, Shaanxi Institute of Ophthalmology, Xi'an, Shaanxi 710002, China
| | - Hongbing Zhang
- Shaanxi Provincial Clinical Research Center for Ophthalmology Diseases, the First Affiliated Hospital of Northwest University, Xi'an No.1 hospital, Xi'an, Shaanxi, China; Shaanxi Key Laboratory of Ophthalmology, Shaanxi Institute of Ophthalmology, Xi'an, Shaanxi 710002, China.
| |
Collapse
|
20
|
Miron RJ, Zhang Y. Understanding exosomes: Part 1-Characterization, quantification and isolation techniques. Periodontol 2000 2024; 94:231-256. [PMID: 37740431 DOI: 10.1111/prd.12520] [Citation(s) in RCA: 24] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 07/21/2023] [Accepted: 07/25/2023] [Indexed: 09/24/2023]
Abstract
Exosomes are the smallest subset of extracellular signaling vesicles secreted by most cells with a diameter in the range of 30-150 nm. Their use has gained great momentum recently due to their ability to be utilized as diagnostic tools with a vast array of therapeutic applications. Over 5000 publications are currently being published yearly on this topic, and this number is only expected to dramatically increase as novel therapeutic strategies continue to be investigated. This review article first focuses on understanding exosomes, including their cellular origin, biogenesis, function, and characterization. Thereafter, overviews of the quantification methods and isolation techniques are given with discussion over their potential use as novel therapeutics in regenerative medicine.
Collapse
Affiliation(s)
- Richard J Miron
- Department of Periodontology, University of Bern, Bern, Switzerland
| | - Yufeng Zhang
- Department of Oral Implantology, University of Wuhan, Wuhan, China
| |
Collapse
|
21
|
van de Wakker SI, Bauzá‐Martinez J, Ríos Arceo C, Manjikian H, Snijders Blok CJB, Roefs MT, Willms E, Maas RGC, Pronker MF, de Jong OG, Wu W, Görgens A, El Andaloussi S, Sluijter JPG, Vader P. Size matters: Functional differences of small extracellular vesicle subpopulations in cardiac repair responses. J Extracell Vesicles 2024; 13:e12396. [PMID: 38179654 PMCID: PMC10767609 DOI: 10.1002/jev2.12396] [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: 07/18/2023] [Accepted: 11/22/2023] [Indexed: 01/06/2024] Open
Abstract
Cardiac progenitor cell (CPC)-derived small extracellular vesicles (sEVs) exhibit great potential to stimulate cardiac repair. However, the multifaceted nature of sEV heterogeneity presents a challenge in understanding the distinct mechanisms underlying their regenerative abilities. Here, a dual-step multimodal flowthrough and size-exclusion chromatography method was applied to isolate and separate CPC-derived sEV subpopulations to study the functional differences related to cardiac repair responses. Three distinct sEV subpopulations were identified with unique protein profiles. Functional cell assays for cardiac repair-related processes demonstrated that the middle-sized and smallest-sized sEV subpopulations exhibited the highest pro-angiogenic and anti-fibrotic activities. Proteasome activity was uniquely seen in the smallest-sized subpopulation. The largest-sized subpopulation showed no effect in any of the functional assays. This research uncovers the existence of sEV subpopulations, each characterized by a distinct composition and biological function. Enhancing our understanding of sEV heterogeneity will provide valuable insights into sEV mechanisms of action, ultimately accelerating the translation of sEV therapeutics.
Collapse
Affiliation(s)
- Simonides Immanuel van de Wakker
- Department of Experimental Cardiology, Regenerative Medicine Center Utrecht, Circulatory health Research CenterUniversity Utrecht, University Medical Center UtrechtUtrechtThe Netherlands
| | - Julia Bauzá‐Martinez
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical SciencesUtrecht UniversityUtrechtThe Netherlands
| | - Carla Ríos Arceo
- Department of Experimental Cardiology, Regenerative Medicine Center Utrecht, Circulatory health Research CenterUniversity Utrecht, University Medical Center UtrechtUtrechtThe Netherlands
| | - Herak Manjikian
- Department of Experimental Cardiology, Regenerative Medicine Center Utrecht, Circulatory health Research CenterUniversity Utrecht, University Medical Center UtrechtUtrechtThe Netherlands
| | - Christian Jamie Bernard Snijders Blok
- Department of Experimental Cardiology, Regenerative Medicine Center Utrecht, Circulatory health Research CenterUniversity Utrecht, University Medical Center UtrechtUtrechtThe Netherlands
| | - Marieke Theodora Roefs
- Department of Experimental Cardiology, Regenerative Medicine Center Utrecht, Circulatory health Research CenterUniversity Utrecht, University Medical Center UtrechtUtrechtThe Netherlands
| | - Eduard Willms
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular ScienceLa Trobe UniversityMelbourneAustralia
| | - Renee Goverdina Catharina Maas
- Department of Experimental Cardiology, Regenerative Medicine Center Utrecht, Circulatory health Research CenterUniversity Utrecht, University Medical Center UtrechtUtrechtThe Netherlands
| | - Matti Feije Pronker
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical SciencesUtrecht UniversityUtrechtThe Netherlands
| | - Olivier Gerrit de Jong
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS)Utrecht UniversityUtrechtThe Netherlands
| | - Wei Wu
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical SciencesUtrecht UniversityUtrechtThe Netherlands
- Singapore Immunology Network (SIgN), Agency for ScienceTechnology and Research (A*STAR)SingaporeSingapore
- Department of PharmacyNational University of SingaporeSingaporeSingapore
| | - André Görgens
- Department of Laboratory MedicineKarolinska InstituteStockholm, HuddingeSweden
- Institute for Transfusion Medicine, University Hospital EssenUniversity of Duisburg‐EssenEssenGermany
| | - Samir El Andaloussi
- Department of Laboratory MedicineKarolinska InstituteStockholm, HuddingeSweden
| | - Joost Petrus Gerardus Sluijter
- Department of Experimental Cardiology, Regenerative Medicine Center Utrecht, Circulatory health Research CenterUniversity Utrecht, University Medical Center UtrechtUtrechtThe Netherlands
| | - Pieter Vader
- Department of Experimental Cardiology, Regenerative Medicine Center Utrecht, Circulatory health Research CenterUniversity Utrecht, University Medical Center UtrechtUtrechtThe Netherlands
- CDL ResearchUniversity Medical Center UtrechtUtrechtThe Netherlands
| |
Collapse
|
22
|
Zheng L, Gong H, Zhang J, Guo L, Zhai Z, Xia S, Hu Z, Chang J, Jiang Y, Huang X, Ge J, Zhang B, Yan M. Strategies to improve the therapeutic efficacy of mesenchymal stem cell-derived extracellular vesicle (MSC-EV): a promising cell-free therapy for liver disease. Front Bioeng Biotechnol 2023; 11:1322514. [PMID: 38155924 PMCID: PMC10753838 DOI: 10.3389/fbioe.2023.1322514] [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: 10/17/2023] [Accepted: 11/29/2023] [Indexed: 12/30/2023] Open
Abstract
Liver disease has emerged as a significant worldwide health challenge due to its diverse causative factors and therapeutic complexities. The majority of liver diseases ultimately progress to end-stage liver disease and liver transplantation remains the only effective therapy with the limitations of donor organ shortage, lifelong immunosuppressants and expensive treatment costs. Numerous pre-clinical studies have revealed that extracellular vesicles released by mesenchymal stem cells (MSC-EV) exhibited considerable potential in treating liver diseases. Although natural MSC-EV has many potential advantages, some characteristics of MSC-EV, such as heterogeneity, uneven therapeutic effect, and rapid clearance in vivo constrain its clinical translation. In recent years, researchers have explored plenty of ways to improve the therapeutic efficacy and rotation rate of MSC-EV in the treatment of liver disease. In this review, we summarized current strategies to enhance the therapeutic potency of MSC-EV, mainly including optimization culture conditions in MSC or modifications of MSC-EV, aiming to facilitate the development and clinical application of MSC-EV in treating liver disease.
Collapse
Affiliation(s)
- Lijuan Zheng
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Institute of Clinical Pharmacy, Central South University, Changsha, China
| | - Hui Gong
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Institute of Clinical Pharmacy, Central South University, Changsha, China
- International Research Center for Precision Medicine, Transformative Technology and Software Services, Changsha, China
| | - Jing Zhang
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
| | - Linna Guo
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Institute of Clinical Pharmacy, Central South University, Changsha, China
- International Research Center for Precision Medicine, Transformative Technology and Software Services, Changsha, China
| | - Zhuofan Zhai
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Shuang Xia
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Institute of Clinical Pharmacy, Central South University, Changsha, China
- International Research Center for Precision Medicine, Transformative Technology and Software Services, Changsha, China
| | - Zhiyu Hu
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Institute of Clinical Pharmacy, Central South University, Changsha, China
- International Research Center for Precision Medicine, Transformative Technology and Software Services, Changsha, China
| | - Jing Chang
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Yizhu Jiang
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Xinran Huang
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Jingyi Ge
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Bikui Zhang
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Institute of Clinical Pharmacy, Central South University, Changsha, China
- International Research Center for Precision Medicine, Transformative Technology and Software Services, Changsha, China
| | - Miao Yan
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Institute of Clinical Pharmacy, Central South University, Changsha, China
- International Research Center for Precision Medicine, Transformative Technology and Software Services, Changsha, China
| |
Collapse
|
23
|
Chen S, Wang Z, Lu H, Yang R, Wu J. Crucial Factors Influencing the Involvement of Odontogenic Exosomes in Dental Pulp Regeneration. Stem Cell Rev Rep 2023; 19:2632-2649. [PMID: 37578647 DOI: 10.1007/s12015-023-10597-z] [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] [Accepted: 07/16/2023] [Indexed: 08/15/2023]
Abstract
Recent progress in exosome based studies has revealed that they possess several advantages over cells, including "cell-free" properties, low immunogenicity and ethical controversy, high biological safety and effective action. These characteristics confer exosomes significant advantages that allow them to overcome the limitations associated with traditional "cell therapy" by circumventing the issues of immune rejection, scarcity of donor cells, heterogeneity, and ethical concerns. Identification of a complete and effective radical treatment for irreversible pulpal disease, a common clinical problem, continues to pose challenges. Although traditional root canal therapy remains the primary clinical treatment, it does not fully restore the physiological functions of pulp. Although stem cell transplantation appears to be a relatively viable treatment strategy for pulp disease, issues such as cell heterogeneity and poor regeneration effects remain problematic. Dental pulp regeneration strategies based on "cell-free" exosome therapies explored by numerous studies appear to have shown significant advantages. In particular, exosomes derived from odontogenic stem cells have demonstrated considerable potential in tooth tissue regeneration engineering, and continue to exhibit superior therapeutic effects compared to non-odontogenic stem cell-derived exosomes. However, only a few studies have comprehensively summarised their research results, particularly regarding the critical factors involved in the process. Therefore, in this study, our purpose was to review the effects exerted by odontogenic exosomes on pulp regeneration and to analyse and discus crucial factors related to this process, thereby providing scholars with a feasible and manageable new concept with respect to regeneration schemes.
Collapse
Affiliation(s)
- San Chen
- Department of Endodontics, School of Stomatology/Affiliated Stomatological Hospital, Zunyi Medical University, Zunyi, 563000, Guizhou, China
| | - Zijie Wang
- Department of Endodontics, School of Stomatology/Affiliated Stomatological Hospital, Zunyi Medical University, Zunyi, 563000, Guizhou, China
| | - Hongqiao Lu
- Department of Endodontics, School of Stomatology/Affiliated Stomatological Hospital, Zunyi Medical University, Zunyi, 563000, Guizhou, China
| | - Runze Yang
- Department of Endodontics, School of Stomatology/Affiliated Stomatological Hospital, Zunyi Medical University, Zunyi, 563000, Guizhou, China
| | - Jiayuan Wu
- Department of Endodontics, School of Stomatology/Affiliated Stomatological Hospital, Zunyi Medical University, Zunyi, 563000, Guizhou, China.
| |
Collapse
|
24
|
Chen S, Wang X, Qian Z, Wang M, Zhang F, Zeng T, Li L, Gao L. Exosomes from ADSCs ameliorate nerve damage in the hippocampus caused by post traumatic brain injury via the delivery of circ-Scmh1 promoting microglial M2 polarization. Injury 2023; 54:110927. [PMID: 37544863 DOI: 10.1016/j.injury.2023.110927] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Revised: 06/21/2023] [Accepted: 07/03/2023] [Indexed: 08/08/2023]
Abstract
BACKGROUND Traumatic brain injury (TBI) is an urgent global health issue. Neuroinflammation, due partially to microglia, can worsen or even cause neuropsychiatric disorders after a TBI. An increasing number of studies have found that adipose-derived stem cell (ADSC) derived exosomes can alleviate many diseases by delivering non-coding RNAs including circRNA and miRNAs, but the mechanism of action remains unclear. METHODS In the present investigation, we produced a TBI mouse model and isolated exosomes from their ADSCs before and after an hypoxic pretreatment. We then used next generation sequencing (NGS) to identify differentially expressed circRNAs and luciferase report assays to determine the relationship between the different noncoding RNAs (miRNA, circRNA and mRNA). RESULTS The results show that we successfully isolated ADSCs which possessed a multidirectional differentiation potential. We then isolated exosomes from untreated ADSCs (Exos) and from hypoxia pretreated ADSCs (HExos). The HExos significantly decreased hippocampal nerve injury after TBI by decreasing M1 microglia mediated inflammatory cytokine expression and caused recovery of cognitive function. NGS data revealed that abnormal circ-Scmh1 expression plays a role in HExo mediated brain tissue preservation after TBI. Furthermore, luciferase report analysis found that miR-154-5p and STAT6 were the targets for circ-Scmh1. Interestingly, miR-154-5p overexpression or STAT6 inhibition reversed the circ-Scmh1 induced M2 microglial polarization. Overexpression of circ-Scmh1 increased the therapeutic effect of Exo on hippocampal nerve injury after TBI by promotion of M2 microglial polarization and decreased inflammatory induced hippocampal nerve injury. CONCLUSION Taken together, we found that exosomes from ADSCs ameliorate nerve damage in the hippocampus post TBI through the delivery of circ-Scmh1 and the promotion of microglial M2 polarization.
Collapse
Affiliation(s)
- Songyu Chen
- Department of Neurosurgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 301 Middle Yanchang Road, Shanghai, 200072, China
| | - Xuewei Wang
- Department of Neurosurgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 301 Middle Yanchang Road, Shanghai, 200072, China
| | - Zhouqi Qian
- Department of Neurosurgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 301 Middle Yanchang Road, Shanghai, 200072, China
| | - Mingsheng Wang
- Department of Neurosurgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 301 Middle Yanchang Road, Shanghai, 200072, China
| | - Feng Zhang
- Department of Neurosurgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 301 Middle Yanchang Road, Shanghai, 200072, China
| | - Tao Zeng
- Department of Neurosurgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 301 Middle Yanchang Road, Shanghai, 200072, China
| | - Lei Li
- Department of Neurosurgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 301 Middle Yanchang Road, Shanghai, 200072, China.
| | - Liang Gao
- Department of Neurosurgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 301 Middle Yanchang Road, Shanghai, 200072, China
| |
Collapse
|
25
|
van de Wakker SI, Meijers FM, Sluijter JPG, Vader P. Extracellular Vesicle Heterogeneity and Its Impact for Regenerative Medicine Applications. Pharmacol Rev 2023; 75:1043-1061. [PMID: 37280097 DOI: 10.1124/pharmrev.123.000841] [Citation(s) in RCA: 46] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 04/20/2023] [Accepted: 05/01/2023] [Indexed: 06/08/2023] Open
Abstract
Extracellular vesicles (EVs) are cell-derived membrane-enclosed particles that are involved in physiologic and pathologic processes. EVs are increasingly being studied for therapeutic applications in the field of regenerative medicine. Therapeutic application of stem cell-derived EVs has shown great potential to stimulate tissue repair. However, the exact mechanisms through which they induce this effect have not been fully clarified. This may to a large extent be attributed to a lack of knowledge on EV heterogeneity. Recent studies suggest that EVs represent a heterogeneous population of vesicles with distinct functions. The heterogeneity of EVs can be attributed to differences in their biogenesis, and as such, they can be classified into distinct populations that can then be further subcategorized into various subpopulations. A better understanding of EV heterogeneity is crucial for elucidating their mechanisms of action in tissue regeneration. This review provides an overview of the latest insights on EV heterogeneity related to tissue repair, including the different characteristics that contribute to such heterogeneity and the functional differences among EV subtypes. It also sheds light on the challenges that hinder clinical translation of EVs. Additionally, innovative EV isolation techniques for studying EV heterogeneity are discussed. Improved knowledge of active EV subtypes would promote the development of tailored EV therapies and aid researchers in the translation of EV-based therapeutics to the clinic. SIGNIFICANCE STATEMENT: Within this review we discuss the differences in regenerative properties of extracellular vesicle (EV) subpopulations and implications of EV heterogeneity for development of EV-based therapeutics. We aim to provide new insights into which aspects are leading to heterogeneity in EV preparations and stress the importance of EV heterogeneity studies for clinical applications.
Collapse
Affiliation(s)
- Simonides Immanuel van de Wakker
- Department of Experimental Cardiology, University Medical Center Utrecht, Utrecht University, The Netherlands (S.I.V.D.W., F.M.M., J.P.G.S., P.V.) and CDL Research, University Medical Center Utrecht, The Netherlands (P.V.)
| | - Fleur Michelle Meijers
- Department of Experimental Cardiology, University Medical Center Utrecht, Utrecht University, The Netherlands (S.I.V.D.W., F.M.M., J.P.G.S., P.V.) and CDL Research, University Medical Center Utrecht, The Netherlands (P.V.)
| | - Joost Petrus Gerardus Sluijter
- Department of Experimental Cardiology, University Medical Center Utrecht, Utrecht University, The Netherlands (S.I.V.D.W., F.M.M., J.P.G.S., P.V.) and CDL Research, University Medical Center Utrecht, The Netherlands (P.V.)
| | - Pieter Vader
- Department of Experimental Cardiology, University Medical Center Utrecht, Utrecht University, The Netherlands (S.I.V.D.W., F.M.M., J.P.G.S., P.V.) and CDL Research, University Medical Center Utrecht, The Netherlands (P.V.)
| |
Collapse
|
26
|
Qin X, He J, Wang X, Wang J, Yang R, Chen X. The functions and clinical application potential of exosomes derived from mesenchymal stem cells on wound repair: a review of recent research advances. Front Immunol 2023; 14:1256687. [PMID: 37691943 PMCID: PMC10486026 DOI: 10.3389/fimmu.2023.1256687] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 08/07/2023] [Indexed: 09/12/2023] Open
Abstract
Wound repair is a complex problem for both clinical practitioners and scientific investigators. Conventional approaches to wound repair have been associated with several limitations, including prolonged treatment duration, high treatment expenses, and significant economic and psychological strain on patients. Consequently, there is a pressing demand for more efficacious and secure treatment modalities to enhance the existing treatment landscapes. In the field of wound repair, cell-free therapy, particularly the use of mesenchymal stem cell-derived exosomes (MSC-Exos), has made notable advancements in recent years. Exosomes, which are small lipid bilayer vesicles discharged by MSCs, harbor bioactive constituents such as proteins, lipids, microRNA (miRNA), and messenger RNA (mRNA). These constituents facilitate material transfer and information exchange between the cells, thereby regulating their biological functions. This article presents a comprehensive survey of the function and mechanisms of MSC-Exos in the context of wound healing, emphasizing their beneficial impact on each phase of the process, including the regulation of the immune response, inhibition of inflammation, promotion of angiogenesis, advancement of cell proliferation and migration, and reduction of scar formation.
Collapse
Affiliation(s)
- Xinchi Qin
- Zunyi Medical University, Zunyi, China
- Department of Burn Surgery, The First People’s Hospital of Foshan, Foshan, China
| | - Jia He
- Department of Burn Surgery, The First People’s Hospital of Foshan, Foshan, China
| | - Xiaoxiang Wang
- Department of Burn Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Jingru Wang
- Department of Burn Surgery, The First People’s Hospital of Foshan, Foshan, China
| | - Ronghua Yang
- Department of Burn and Plastic Surgery, Guangzhou First People’s Hospital, South China University of Technology, Guangzhou, China
| | - Xiaodong Chen
- Zunyi Medical University, Zunyi, China
- Department of Burn Surgery, The First People’s Hospital of Foshan, Foshan, China
| |
Collapse
|
27
|
Zhang L, Liu J, Zhou C. Current aspects of small extracellular vesicles in pain process and relief. Biomater Res 2023; 27:78. [PMID: 37563666 PMCID: PMC10416402 DOI: 10.1186/s40824-023-00417-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 08/04/2023] [Indexed: 08/12/2023] Open
Abstract
Small extracellular vesicles (sEVs) have been identified as a noteworthy paracrine mechanism of intercellular communication in diagnosing and managing neurological disorders. Current research suggests that sEVs play a pivotal role in the pathological progression of pain, emphasizing their critical function in the pathological progression of pain in acute and chronic pain models. By facilitating the transfer of diverse molecules, such as proteins, nucleic acids, and metabolites, sEVs can modulate pain signaling transmission in both the central and peripheral nervous systems. Furthermore, the unique molecules conveyed by sEVs in pain disorders indicate their potential as diagnostic biomarkers. The application of sEVs derived from mesenchymal stem cells (MSCs) in regenerative pain medicine has emerged as a promising strategy for pain management. Moreover, modified sEVs have garnered considerable attention in the investigation of pathological processes and therapeutic interventions. This review presents a comprehensive overview of the current knowledge regarding the involvement of sEVs in pain pathogenesis and treatment. Nevertheless, additional research is imperative to facilitate their clinical implementation. Schematic diagram of sEVs in the biogenesis, signal transmission, diagnosis, and treatment of pain disorders. Small extracellular vesicles (sEVs) are secreted by multiple cells, loading with various biomolecules, such as miRNAs, transmembrane proteins, and amino acids. They selectively target other cells and regulating pain signal transmission. The composition of sEVs can serve as valuable biomarkers for pain diagnosis. In particular, mesenchymal stem cell-derived sEVs have shown promise as regenerative medicine for managing multiple pain disorders. Furthermore, by modifying the structure or contents of sEVs, they could potentially be used as a potent analgesic method.
Collapse
Affiliation(s)
- Lanyu Zhang
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Anesthesia & Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital of Sichuan University, Chengdu, 610041, Sichuan, China
| | - Jin Liu
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China
| | - Cheng Zhou
- Laboratory of Anesthesia & Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital of Sichuan University, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
28
|
Yan L, Li J, Zhang C. The role of MSCs and CAR-MSCs in cellular immunotherapy. Cell Commun Signal 2023; 21:187. [PMID: 37528472 PMCID: PMC10391838 DOI: 10.1186/s12964-023-01191-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Accepted: 06/07/2023] [Indexed: 08/03/2023] Open
Abstract
Chimeric antigen receptors (CARs) are widely used by T cells (CAR-T cells), natural killer cells dendritic cells and macrophages, and they are of great importance in cellular immunotherapy. However, the use of CAR-related products faces several challenges, including the poor persistence of cells carrying CARs, cell dysfunction or exhaustion, relapse of disease, immune effector cell-associated neurotoxicity syndrome, cytokine release syndrome, low efficacy against solid tumors and immunosuppression by the tumor microenvironment. Another important cell therapy regimen involves mesenchymal stem cells (MSCs). Recent studies have shown that MSCs can improve the anticancer functions of CAR-related products. CAR-MSCs can overcome the flaws of cellular immunotherapy. Thus, MSCs can be used as a biological vehicle for CARs. In this review, we first discuss the characteristics and immunomodulatory functions of MSCs. Then, the role of MSCs as a source of exosomes, including the characteristics of MSC-derived exosomes and their immunomodulatory functions, is discussed. The role of MSCs in CAR-related products, CAR-related product-derived exosomes and the effect of MSCs on CAR-related products are reviewed. Finally, the use of MSCs as CAR vehicles is discussed. Video Abstract.
Collapse
Affiliation(s)
- Lun Yan
- Medical Center of Hematology, State Key Laboratory of Trauma, Burn and Combined Injury, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China
| | - Jing Li
- Medical Center of Hematology, State Key Laboratory of Trauma, Burn and Combined Injury, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China
| | - Cheng Zhang
- Medical Center of Hematology, State Key Laboratory of Trauma, Burn and Combined Injury, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China.
| |
Collapse
|
29
|
de Pedro MÁ, López E, González-Nuño FM, Pulido M, Álvarez V, Marchena AM, Preußer C, Szymański W, Pogge von Strandmann E, Graumann J, Sánchez-Margallo FM, Casado JG, Gómez-Serrano M. Menstrual blood-derived mesenchymal stromal cells: impact of preconditioning on the cargo of extracellular vesicles as potential therapeutics. Stem Cell Res Ther 2023; 14:187. [PMID: 37507751 PMCID: PMC10386225 DOI: 10.1186/s13287-023-03413-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 07/12/2023] [Indexed: 07/30/2023] Open
Abstract
BACKGROUND Mesenchymal stromal cells (MSCs) have been shown to exert their therapeutic effects through the secretion of broad spectrum of paracrine factors, including extracellular vesicles (EVs). Accordingly, EVs are being pursued as a promising alternative to cell-based therapies. Menstrual blood-derived stromal cells (MenSCs) are a type of MSC that, due to their immunomodulatory and regenerative properties, have emerged as an innovative source. Additionally, new strategies of cell priming may potentially alter the concentration and cargo of released EVs, leading to modification of their biological properties. In this study, we aimed to characterize the EVs released by MenSCs and compare their therapeutic potential under three different preconditioning conditions (proinflammatory stimuli, physioxia, and acute hypoxia). METHODS MenSCs were isolated from five healthy women. Following culturing to 80% confluence, MenSCs were exposed to different priming conditions: basal (21% O2), proinflammatory stimuli (IFNγ and TNFα, 21% O2), physioxia (1-2% O2), and acute hypoxia (< 1% O2) for 48-72 h. Conditioned media from MenSCs was collected after 48 h and EVs were isolated by a combination of ultra-filtration and differential centrifugation. An extensive characterization ranging from nano-flow cytometry (nFC) to quantitative high-throughput shotgun proteomics was performed. Bioinformatics analyses were used to derive hypotheses on their biological properties. RESULTS No differences in the morphology, size, or number of EVs released were detected between priming conditions. The proteome analysis associated with basal MenSC-EVs prominently revealed their immunomodulatory and regenerative capabilities. Furthermore, quantitative proteomic analysis of differentially produced MenSC-EVs provided sufficient evidence for the utility of the differential preconditioning in purpose-tailoring EVs for their therapeutic application: proinflammatory priming enhanced the anti-inflammatory, regenerative and immunomodulatory capacity in the innate response of EVs, physioxia priming also improves tissue regeneration, angiogenesis and their immunomodulatory capacity targeting on the adaptive response, while acute hypoxia priming, increased hemostasis and apoptotic processes regulation in MenSC-EVs, also by stimulating immunomodulation mainly through the adaptive response. CONCLUSIONS Priming of MenSCs under proinflammatory and hypoxic conditions affected the cargo proteome of EVs released, resulting in different therapeutic potential, and thus warrants experimental exploration with the aim to generate better-defined MSC-derived bioproducts.
Collapse
Affiliation(s)
- María Ángeles de Pedro
- Stem Cell Therapy Unit, Jesús Usón Minimally Invasive Surgery Centre, 10071, Cáceres, Spain
- RICORS-TERAV Network, ISCIII, 28029, Madrid, Spain
| | - Esther López
- Stem Cell Therapy Unit, Jesús Usón Minimally Invasive Surgery Centre, 10071, Cáceres, Spain.
- RICORS-TERAV Network, ISCIII, 28029, Madrid, Spain.
| | | | - María Pulido
- Stem Cell Therapy Unit, Jesús Usón Minimally Invasive Surgery Centre, 10071, Cáceres, Spain
| | - Verónica Álvarez
- Stem Cell Therapy Unit, Jesús Usón Minimally Invasive Surgery Centre, 10071, Cáceres, Spain
| | - Ana María Marchena
- Stem Cell Therapy Unit, Jesús Usón Minimally Invasive Surgery Centre, 10071, Cáceres, Spain
- RICORS-TERAV Network, ISCIII, 28029, Madrid, Spain
| | - Christian Preußer
- Institute for Tumor Immunology, Center for Tumor Biology and Immunology, Philipps University, 35043, Marburg, Germany
- Core Facility Extracellular Vesicles, Center for Tumor Biology and Immunology, Philipps University, 35043, Marburg, Germany
| | - Witold Szymański
- Institute of Translational Proteomics, Biochemical/Pharmacological Center, Philipps University, 35043, Marburg, Germany
| | - Elke Pogge von Strandmann
- Institute for Tumor Immunology, Center for Tumor Biology and Immunology, Philipps University, 35043, Marburg, Germany
- Core Facility Extracellular Vesicles, Center for Tumor Biology and Immunology, Philipps University, 35043, Marburg, Germany
| | - Johannes Graumann
- Institute of Translational Proteomics, Biochemical/Pharmacological Center, Philipps University, 35043, Marburg, Germany
| | - Francisco Miguel Sánchez-Margallo
- Stem Cell Therapy Unit, Jesús Usón Minimally Invasive Surgery Centre, 10071, Cáceres, Spain
- RICORS-TERAV Network, ISCIII, 28029, Madrid, Spain
| | - Javier G Casado
- RICORS-TERAV Network, ISCIII, 28029, Madrid, Spain
- Immunology Unit, University of Extremadura, 10003, Cáceres, Spain
- Institute of Molecular Pathology Biomarkers, University of Extremadura, 10003, Cáceres, Spain
| | - María Gómez-Serrano
- Institute for Tumor Immunology, Center for Tumor Biology and Immunology, Philipps University, 35043, Marburg, Germany.
| |
Collapse
|
30
|
Lee DH, Yun DW, Kim YH, Im GB, Hyun J, Park HS, Bhang SH, Choi SH. Various Three-Dimensional Culture Methods and Cell Types for Exosome Production. Tissue Eng Regen Med 2023; 20:621-635. [PMID: 37269439 PMCID: PMC10313642 DOI: 10.1007/s13770-023-00551-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 04/06/2023] [Accepted: 05/02/2023] [Indexed: 06/05/2023] Open
Abstract
Cell-based therapies have been used as promising treatments for several untreatable diseases. However, cell-based therapies have side effects such as tumorigenesis and immune responses. To overcome these side effects, therapeutic effects of exosomes have been researched as replacements for cell-based therapies. In addition, exosomes reduced the risk that can be induced by cell-based therapies. Exosomes contain biomolecules such as proteins, lipids, and nucleic acids that play an essential role in cell-cell and cell-matrix interactions during biological processes. Since the introduction of exosomes, those have been proven perpetually as one of the most effective and therapeutic methods for incurable diseases. Much research has been conducted to enhance the properties of exosomes, including immune regulation, tissue repair, and regeneration. However, yield rate of exosomes is the critical obstacle that should be overcome for practical cell-free therapy. Three-dimensional (3D) culture methods are introduced as a breakthrough to get higher production yields of exosomes. For example, hanging drop and microwell were well known 3D culture methods and easy to use without invasiveness. However, these methods have limitation in mass production of exosomes. Therefore, a scaffold, spinner flask, and fiber bioreactor were introduced for mass production of exosomes isolated from various cell types. Furthermore, exosomes treatments derived from 3D cultured cells showed enhanced cell proliferation, angiogenesis, and immunosuppressive properties. This review provides therapeutic applications of exosomes using 3D culture methods.
Collapse
Affiliation(s)
- Dong-Hyun Lee
- School of Chemical Engineering, Sungkyunkwan University (SKKU), 2066 Seobu-Ro, Jangan-Gu, Suwon, Gyeonggi-Do, 16419, Republic of Korea
| | - Dae Won Yun
- School of Chemical Engineering, Sungkyunkwan University (SKKU), 2066 Seobu-Ro, Jangan-Gu, Suwon, Gyeonggi-Do, 16419, Republic of Korea
| | - Yeong Hwan Kim
- School of Chemical Engineering, Sungkyunkwan University (SKKU), 2066 Seobu-Ro, Jangan-Gu, Suwon, Gyeonggi-Do, 16419, Republic of Korea
| | - Gwang-Bum Im
- School of Chemical Engineering, Sungkyunkwan University (SKKU), 2066 Seobu-Ro, Jangan-Gu, Suwon, Gyeonggi-Do, 16419, Republic of Korea
| | - Jiyu Hyun
- School of Chemical Engineering, Sungkyunkwan University (SKKU), 2066 Seobu-Ro, Jangan-Gu, Suwon, Gyeonggi-Do, 16419, Republic of Korea
| | - Hyun Su Park
- School of Chemical Engineering, Sungkyunkwan University (SKKU), 2066 Seobu-Ro, Jangan-Gu, Suwon, Gyeonggi-Do, 16419, Republic of Korea
| | - Suk Ho Bhang
- School of Chemical Engineering, Sungkyunkwan University (SKKU), 2066 Seobu-Ro, Jangan-Gu, Suwon, Gyeonggi-Do, 16419, Republic of Korea.
| | - Sang Hyoun Choi
- Department of Radiation Oncology, Korea Institute of Radiological and Medical Science, Seoul, Republic of Korea.
| |
Collapse
|
31
|
Pincela Lins PM, Pirlet E, Szymonik M, Bronckaers A, Nelissen I. Manufacture of extracellular vesicles derived from mesenchymal stromal cells. Trends Biotechnol 2023; 41:965-981. [PMID: 36750391 DOI: 10.1016/j.tibtech.2023.01.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 12/09/2022] [Accepted: 01/05/2023] [Indexed: 02/08/2023]
Abstract
Mesenchymal stromal cells (MSCs) are a promising therapy for various diseases ranging from ischemic stroke to wound healing and cancer. Their therapeutic effects are mainly mediated by secretome-derived paracrine factors, with extracellular vesicles (EVs) proven to play a key role. This has led to promising research on the potential of MSC-EVs as regenerative, off-the-shelf therapeutic agents. However, the translation of MSC-EVs into the clinic is hampered by the poor scalability of their production. Recently, new advanced methods have been developed to upscale MSC cultivation and EV production yields, ranging from new cell culture devices to priming procedures. This review gives an overview of these innovative strategies for manufacturing MSC-EVs.
Collapse
Affiliation(s)
- Paula M Pincela Lins
- Hasselt University, Faculty of Medicine and Life Sciences, Biomedical Research Institute (BIOMED), Agoralaan, 3590 Diepenbeek, Belgium; Flemish Institute for Technological Research (VITO), Health Department, Boeretang, 2400 Mol, Belgium
| | - Elke Pirlet
- Hasselt University, Faculty of Medicine and Life Sciences, Biomedical Research Institute (BIOMED), Agoralaan, 3590 Diepenbeek, Belgium
| | - Michal Szymonik
- Flemish Institute for Technological Research (VITO), Health Department, Boeretang, 2400 Mol, Belgium
| | - Annelies Bronckaers
- Hasselt University, Faculty of Medicine and Life Sciences, Biomedical Research Institute (BIOMED), Agoralaan, 3590 Diepenbeek, Belgium.
| | - Inge Nelissen
- Flemish Institute for Technological Research (VITO), Health Department, Boeretang, 2400 Mol, Belgium.
| |
Collapse
|
32
|
Yang M, Peng GH. The molecular mechanism of human stem cell-derived extracellular vesicles in retinal repair and regeneration. Stem Cell Res Ther 2023; 14:84. [PMID: 37046324 PMCID: PMC10100447 DOI: 10.1186/s13287-023-03319-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Accepted: 03/29/2023] [Indexed: 04/14/2023] Open
Abstract
Extracellular vesicles (EVs), including microvesicles (MVs) and exosomes, play a critical role in metabolic regulation and intracellular communication. Stem cell-derived EVs are considered to have the potential for regeneration, like stem cells, while simultaneously avoiding the risk of immune rejection or tumour formation. The therapeutic effect of stem cell-derived EVs has been proven in many diseases. However, the molecular mechanism of stem cell-derived EVs in retinal repair and regeneration has not been fully clarified. In this review, we described the biological characteristics of stem cell-derived EVs, summarized the current research on stem cell-derived EV treatment in retinal repair and regeneration, and discussed the potential and challenges of stem cell-derived EVs in translational medicine.
Collapse
Affiliation(s)
- Mei Yang
- Laboratory of Visual Cell Differentiation and Regulation, Basic Medical College, Zhengzhou University, Zhengzhou, 450001, China
- Department of Pathophysiology, Basic Medical College, Zhengzhou University, Zhengzhou, 450001, China
| | - Guang-Hua Peng
- Laboratory of Visual Cell Differentiation and Regulation, Basic Medical College, Zhengzhou University, Zhengzhou, 450001, China.
- Department of Pathophysiology, Basic Medical College, Zhengzhou University, Zhengzhou, 450001, China.
| |
Collapse
|
33
|
Sarasati A, Syahruddin MH, Nuryanti A, Ana ID, Barlian A, Wijaya CH, Ratnadewi D, Wungu TDK, Takemori H. Plant-Derived Exosome-like Nanoparticles for Biomedical Applications and Regenerative Therapy. Biomedicines 2023; 11:biomedicines11041053. [PMID: 37189671 DOI: 10.3390/biomedicines11041053] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 03/07/2023] [Accepted: 03/09/2023] [Indexed: 03/31/2023] Open
Abstract
Plant-derived exosome-like nanoparticles (PDENs) comprise various bioactive biomolecules. As an alternative cell-free therapeutic approach, they have the potential to deliver nano-bioactive compounds to the human body, and thus lead to various anti-inflammatory, antioxidant, and anti-tumor benefits. Moreover, it is known that Indonesia is one of the herbal centers of the world, with an abundance of unexplored sources of PDENs. This encouraged further research in biomedical science to develop natural richness in plants as a source for human welfare. This study aims to verify the potential of PDENs for biomedical purposes, especially for regenerative therapy applications, by collecting and analyzing data from the latest relevant research and developments.
Collapse
|
34
|
Zhang X, Jiang W, Lu Y, Mao T, Gu Y, Ju D, Dong C. Exosomes combined with biomaterials in the treatment of spinal cord injury. Front Bioeng Biotechnol 2023; 11:1077825. [PMID: 36994357 PMCID: PMC10040754 DOI: 10.3389/fbioe.2023.1077825] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Accepted: 02/27/2023] [Indexed: 03/16/2023] Open
Abstract
Spinal cord injury (SCI) is a serious and disabling disease with a high mortality rate. It often leads to complete or partial sensory and motor dysfunction and is accompanied by a series of secondary outcomes, such as pressure sores, pulmonary infections, deep vein thrombosis in the lower extremities, urinary tract infections, and autonomic dysfunction. Currently, the main treatments for SCI include surgical decompression, drug therapy, and postoperative rehabilitation. Studies have shown that cell therapy plays a beneficial role in the treatment of SCI. Nonetheless, there is controversy regarding the therapeutic effect of cell transplantation in SCI models. Meanwhile exosomes, as a new therapeutic medium for regenerative medicine, possess the advantages of small size, low immunogenicity, and the ability to cross the blood-spinal cord barrier. Certain studies have shown that stem cell-derived exosomes have anti-inflammatory effects and can play an irreplaceable role in the treatment of SCI. In this case, it is difficult for a single treatment method to play an effective role in the repair of neural tissue after SCI. The combination of biomaterial scaffolds and exosomes can better transfer and fix exosomes to the injury site and improve their survival rate. This paper first reviews the current research status of stem cell-derived exosomes and biomaterial scaffolds in the treatment of SCI respectively, and then describes the application of exosomes combined with biomaterial scaffolds in the treatment of SCI, as well as the challenges and prospects.
Collapse
|
35
|
Geng Z, Guo H, Li Y, Liu Y, Zhao Y. Stem cell-derived extracellular vesicles: A novel and potential remedy for primary ovarian insufficiency. Front Cell Dev Biol 2023; 11:1090997. [PMID: 36875770 PMCID: PMC9977284 DOI: 10.3389/fcell.2023.1090997] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Accepted: 02/06/2023] [Indexed: 02/17/2023] Open
Abstract
Primary ovarian insufficiency (POI) is an essential cause of young female fertility loss. At present, there are many treatments for primary ovarian insufficiency, but due to the complexity of the pathogenesis of primary ovarian insufficiency, the efficacy still could not be satisfactory. Stem cell transplantation is a feasible intervention protocol for primary ovarian insufficiency. However, its wide application in the clinic is limited by some defects such as tumorigenic and controversial ethical issues. Stem cell-derived extracellular vesicles (EVs) represent an important mode of intercellular communication attracting increasing interest. It is well documented that stem cell-derived extracellular vesicles for primary ovarian insufficiency with exciting therapeutic effects. Studies have found that stem cell-derived extracellular vesicles could improve ovarian reserve, increase the growth of follicles, reduce follicle atresia, and restore hormone levels of FSH and E2. Its mechanisms include inhibiting ovarian granulosa cells (GCs) apoptosis, reactive oxygen species, and inflammatory response and promoting granulosa cells proliferation and angiogenesis. Thus, stem cell-derived extracellular vesicles are a promising and potential method for primary ovarian insufficiency patients. However, stem cell-derived extracellular vesicles are still a long way from clinical translation. This review will provide an overview of the role and the mechanisms of stem cell-derived extracellular vesicles in primary ovarian insufficiency, and further elaborate on the current challenges. It may suggest new directions for future research.
Collapse
Affiliation(s)
- Zixiang Geng
- Shi’s Center of Orthopedics and Traumatology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Traumatology and Orthopedics, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| | - Hailing Guo
- Shi’s Center of Orthopedics and Traumatology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Traumatology and Orthopedics, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| | - Yifei Li
- Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ying Liu
- Department of Dermatology, Shanghai Songjiang District Central Hospital, Shanghai, China
| | - Yongfang Zhao
- Shi’s Center of Orthopedics and Traumatology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Traumatology and Orthopedics, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
36
|
Exosome-Based Carrier for RNA Delivery: Progress and Challenges. Pharmaceutics 2023; 15:pharmaceutics15020598. [PMID: 36839920 PMCID: PMC9964211 DOI: 10.3390/pharmaceutics15020598] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Revised: 02/01/2023] [Accepted: 02/02/2023] [Indexed: 02/12/2023] Open
Abstract
In the last few decades, RNA-based drugs have emerged as a promising candidate to specifically target and modulate disease-relevant genes to cure genetic defects. The key to applying RNA therapy in clinical trials is developing safe and effective delivery systems. Exosomes have been exploited as a promising vehicle for drug delivery due to their nanoscale size, high stability, high biocompatibility, and low immunogenicity. We reviewed and summarized the progress in the strategy and application of exosome-mediated RNA therapy. The challenges of exosomes as a carrier for RNA drug delivery are also elucidated in this article. RNA molecules can be loaded into exosomes and then delivered to targeted cells or tissues via various biochemical or physical approaches. So far, exosome-mediated RNA therapy has shown potential in the treatment of cancer, central nervous system disorders, COVID-19, and other diseases. To further exploit the potential of exosomes for RNA delivery, more efforts should be made to overcome both technological and logistic problems.
Collapse
|
37
|
Rehman FU, Liu Y, Zheng M, Shi B. Exosomes based strategies for brain drug delivery. Biomaterials 2023; 293:121949. [PMID: 36525706 DOI: 10.1016/j.biomaterials.2022.121949] [Citation(s) in RCA: 99] [Impact Index Per Article: 49.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 11/12/2022] [Accepted: 12/05/2022] [Indexed: 12/13/2022]
Abstract
Exosome application has emerged as a promising nanotechnology discipline for various diseases therapeutics and diagnoses. Owing to the natural properties of efficient drug delivery, higher biocompatibility, facile traversing of physiological barriers, and subtle side effects, exosomes shorten their way to clinical translation. Exosomes are nanoscale membrane-bound vesicles primarily involved in intercellular communication and exhibit natural blood-brain barrier (BBB) traversing ability, which enables their application as drug delivery vehicles for brain diseases treatment. Herein, we highlight recent exosome-based drug delivery endeavors for neurodegenerative diseases and brain cancer therapy, summarize the obstacles and future directions in clinical translation.
Collapse
Affiliation(s)
- Fawad Ur Rehman
- Henan-Macquire International Joint Center for Biomedical Innovations, School of Life Sciences, Henan University, JinMing Avenue, Kaifeng, 475004 PR China; Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences & School of Pharmacy, Henan University, Kaifeng, Henan 475004, China; Centre for Regenerative Medicine and Stem Cells Research, The Aga Khan University, Stadium Road, Karachi, 74800, Pakistan
| | - Yang Liu
- Henan-Macquire International Joint Center for Biomedical Innovations, School of Life Sciences, Henan University, JinMing Avenue, Kaifeng, 475004 PR China; Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences & School of Pharmacy, Henan University, Kaifeng, Henan 475004, China
| | - Meng Zheng
- Henan-Macquire International Joint Center for Biomedical Innovations, School of Life Sciences, Henan University, JinMing Avenue, Kaifeng, 475004 PR China; Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences & School of Pharmacy, Henan University, Kaifeng, Henan 475004, China.
| | - Bingyang Shi
- Henan-Macquire International Joint Center for Biomedical Innovations, School of Life Sciences, Henan University, JinMing Avenue, Kaifeng, 475004 PR China; Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences & School of Pharmacy, Henan University, Kaifeng, Henan 475004, China; Department of Biomedical Sciences Faculty of Medicine and Health Sciences Macquarie University Sydney, NSW, 2109, Australia.
| |
Collapse
|
38
|
A Comparative Study of Mesenchymal Stem Cell-Derived Extracellular Vesicles' Local and Systemic Dose-Dependent Administration in Rat Spinal Cord Injury. BIOLOGY 2022; 11:biology11121853. [PMID: 36552362 PMCID: PMC9775578 DOI: 10.3390/biology11121853] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 12/09/2022] [Accepted: 12/15/2022] [Indexed: 12/23/2022]
Abstract
Spinal cord injury (SCI) is a serious neurological condition that causes severe disability. One of the approaches to overcoming the complications of SCI is stem cell-derived extracellular vesicle (EV) therapy. In this research, we performed a comparative evaluation of rat spinal cord post-traumatic regeneration efficacy using different methods of mesenchymal stem cell-derived EV transplantation (local vs. systemic) followed by evaluation of their minimal therapeutic dose. The results suggested that MSC-EV therapy could improve locomotor activity over 60 days after the SCI, showing a dose-dependent effect on the recovery of spinal cord motor pathways. We also established the possibility of maintaining a population of mature oligodendrocytes by MSC-EVs. It was observed that in the spinal cord injury area, intravenous transplantation of MSC-EVs showed more pronounced therapeutic effects compared to the treatment of fibrin matrix-encapsulated MSC-EVs.
Collapse
|
39
|
Luo Z, Sun Y, Qi B, Lin J, Chen Y, Xu Y, Chen J. Human bone marrow mesenchymal stem cell-derived extracellular vesicles inhibit shoulder stiffness via let-7a/Tgfbr1 axis. Bioact Mater 2022; 17:344-359. [PMID: 35386460 PMCID: PMC8965035 DOI: 10.1016/j.bioactmat.2022.01.016] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 12/30/2021] [Accepted: 01/09/2022] [Indexed: 02/07/2023] Open
Abstract
Shoulder stiffness (SS) is a common shoulder disease characterized by increasing pain and limited range of motion. SS is considered to be an inflammatory and fibrotic disorder pathologically. However, there is no consensus on the most effective conservative treatment for fibrosis. Given that human Bone Marrow Mesenchymal Stem Cell-derived extracellular vesicles (BMSC-EVs) displayed promising therapeutic effects for various tissues, we investigated the therapeutic effect of BMSC-EVs on fibrosis in a mice immobilization model and two cell models. By conducting a series of experiments, we found that BMSC-EVs can significantly inhibit the fibrogenic process both in vitro and in vivo. In detail, BMSC-EVs suppressed the aberrant proliferation, high collagen production capacity, and activation of fibrotic pathways in TGF-β-stimulated fibroblasts in vitro. Besides, in vivo, BMSC-EVs reduced cell infiltration, reduced fibrotic tissue in the shoulder capsule, and improved shoulder mobility. In addition, via exosomal small RNA sequencing and qPCR analysis, let-7a-5p was verified to be the highest expressed miRNA with predicted antifibrotic capability in BMSC-EVs. The antifibrotic capacity of BMSC-EVs was significantly impaired after the knockdown of let-7a-5p. Moreover, we discovered that the mRNA of TGFBR1 (the membrane receptor of transforming growth factor β) was the target of let-7a-5p. Together, these findings elucidated the antifibrotic role of BMSC-EVs in shoulder capsular fibrosis. This study clarifies a new approach using stem cell-derived EVs therapy as an alternative to cell therapy, which may clinically benefit patients with SS in the future.
Collapse
Affiliation(s)
- Zhiwen Luo
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Yaying Sun
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Beijie Qi
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Jinrong Lin
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Yisheng Chen
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Yuzhen Xu
- Department of Rehabilitation, The Second Affiliated Hospital of Shandong First Medical University, Shandong Province, China
| | - Jiwu Chen
- Department of Sports Medicine, Shanghai General Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
40
|
MSC-EV therapy for bone/cartilage diseases. Bone Rep 2022; 17:101636. [DOI: 10.1016/j.bonr.2022.101636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 11/03/2022] [Accepted: 11/08/2022] [Indexed: 11/11/2022] Open
|
41
|
Composition, Biogenesis, and Role of Exosomes in Tumor Development. Stem Cells Int 2022; 2022:8392509. [PMID: 36117723 PMCID: PMC9481374 DOI: 10.1155/2022/8392509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 08/14/2022] [Accepted: 08/26/2022] [Indexed: 11/30/2022] Open
Abstract
The role of exosomes and their mechanism of action at the tumor site have received increasing attention. These microvesicles are produced by a wide range of cells including mesenchymal stem cells (MSCs) and immune cells. In particular, tumor cells release remarkable amounts of exosomes which spread to distant organs through the blood and enhance the possibility of tumor metastasis. In spite of results on tumor promoting properties, there are reports demonstrating the tumor inhibiting effects of exosomes depending on the type of the tumor and cell source. This review aims to have a comprehensive appraisal on the biogenesis, composition, and isolation of exosomes and then highlights the current knowledge of their role in cancer progression or inhibition by special focusing on MSC's exosomes (MSC-EXOs).
Collapse
|
42
|
Liu X, Li X, Wu G, Qi P, Zhang Y, Liu Z, Li X, Yu Y, Ye X, Li Y, Yang D, Teng Y, Shi C, Jin X, Qi S, Liu Y, Wang S, Liu Y, Cao F, Kong Q, Wang Z, Zhang H. Umbilical Cord Mesenchymal Stem Cell-Derived Small Extracellular Vesicles Deliver miR-21 to Promote Corneal Epithelial Wound Healing through PTEN/PI3K/Akt Pathway. Stem Cells Int 2022; 2022:1252557. [PMID: 35873535 PMCID: PMC9303509 DOI: 10.1155/2022/1252557] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 05/19/2022] [Accepted: 06/23/2022] [Indexed: 12/30/2022] Open
Abstract
Objective Rapid restoration of corneal epithelium integrity after injury is particularly important for preserving corneal transparency and vision. Mesenchymal stem cells (MSCs) can be taken into account as the promising regenerative therapeutics for improvement of wound healing processes based on the variety of the effective components. The extracellular vesicles form MSCs, especially exosomes, have been considered as important paracrine mediators though transferring microRNAs into recipient cell. This study investigated the mechanism of human umbilical cord MSC-derived small extracellular vesicles (HUMSC-sEVs) on corneal epithelial wound healing. Methods HUMSC-sEVs were identified by transmission electron microscopy, nanoparticle tracking analysis, and Western blot. Corneal fluorescein staining and histological staining were evaluated in a corneal mechanical wound model. Changes in HCEC proliferation after HUMSC-sEVs or miR-21 mimic treatment were evaluated by CCK-8 and EdU assays, while migration was assessed by in vitro scratch wound assay. Full-length transcriptome sequencing was performed to identify the differentially expressed genes associated with HUMSC-sEVs treatment, followed by validation via real-time PCR and Western blot. Results The sEVs derived from HUMSCs can significantly promote corneal epithelial cell proliferation, migration in vitro, and corneal epithelial wound healing in vivo. Similar effects were obtained after miR-21 transfection, while the beneficial effects of HUMSC-sEVs were partially negated by miR-21 knockdown. Results also show that the benefits are associated with decreased PTEN level and activated the PI3K/Akt signaling pathway in HCECs. Conclusion HUMSC-sEVs could enhance the recovery of corneal epithelial wounds though restraining PTEN by transferring miR-21 and may represent a promising novel therapeutic agent for corneal wound repair.
Collapse
Affiliation(s)
- Xiaolong Liu
- Central Laboratory, First Affiliated Hospital, Harbin Medical University, Harbin, China
- College of Life Science, Northeast Agricultural University, Harbin, China
| | - Xuran Li
- Eye Hospital, First Affiliated Hospital, Harbin Medical University, Harbin, China
- NHC Key Laboratory of Cell Transplantation, Harbin Medical University, Harbin, China
| | - Guangyuan Wu
- Central Laboratory, First Affiliated Hospital, Harbin Medical University, Harbin, China
- NHC Key Laboratory of Cell Transplantation, Harbin Medical University, Harbin, China
| | - Pengfei Qi
- Central Laboratory, First Affiliated Hospital, Harbin Medical University, Harbin, China
- NHC Key Laboratory of Cell Transplantation, Harbin Medical University, Harbin, China
| | - Yanyan Zhang
- Eye Hospital, First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Zhiyu Liu
- Department of Laboratory Diagnostics, First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Xinyue Li
- Eye Hospital, First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Yu Yu
- Eye Hospital, First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Xiangmei Ye
- Central Laboratory, First Affiliated Hospital, Harbin Medical University, Harbin, China
- NHC Key Laboratory of Cell Transplantation, Harbin Medical University, Harbin, China
| | - Yang Li
- Central Laboratory, First Affiliated Hospital, Harbin Medical University, Harbin, China
- NHC Key Laboratory of Cell Transplantation, Harbin Medical University, Harbin, China
| | - Dongguang Yang
- Central Laboratory, First Affiliated Hospital, Harbin Medical University, Harbin, China
- NHC Key Laboratory of Cell Transplantation, Harbin Medical University, Harbin, China
| | - Yueqiu Teng
- Central Laboratory, First Affiliated Hospital, Harbin Medical University, Harbin, China
- NHC Key Laboratory of Cell Transplantation, Harbin Medical University, Harbin, China
| | - Ce Shi
- Central Laboratory, First Affiliated Hospital, Harbin Medical University, Harbin, China
- NHC Key Laboratory of Cell Transplantation, Harbin Medical University, Harbin, China
| | - Xin Jin
- Eye Hospital, First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Sen Qi
- Department of Hematology, First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Yuting Liu
- Eye Hospital, First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Shudan Wang
- Eye Hospital, First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Ying Liu
- Eye Hospital, First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Fenglin Cao
- Central Laboratory, First Affiliated Hospital, Harbin Medical University, Harbin, China
- NHC Key Laboratory of Cell Transplantation, Harbin Medical University, Harbin, China
| | - Qingran Kong
- College of Life Science, Northeast Agricultural University, Harbin, China
| | - Zhenkun Wang
- Central Laboratory, First Affiliated Hospital, Harbin Medical University, Harbin, China
- NHC Key Laboratory of Cell Transplantation, Harbin Medical University, Harbin, China
| | - Hong Zhang
- Eye Hospital, First Affiliated Hospital, Harbin Medical University, Harbin, China
- NHC Key Laboratory of Cell Transplantation, Harbin Medical University, Harbin, China
| |
Collapse
|
43
|
Macrophage bioassay standardization to assess the anti-inflammatory activity of mesenchymal stromal cell-derived small extracellular vesicles. Cytotherapy 2022; 24:999-1012. [PMID: 35798638 DOI: 10.1016/j.jcyt.2022.05.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 05/23/2022] [Accepted: 05/30/2022] [Indexed: 12/18/2022]
Abstract
BACKGROUND AIMS Owing to the lack of biological assays, determining the biological activity of extracellular vesicles has proven difficult. Here the authors standardized an in vitro assay to assess the anti-inflammatory activity of mesenchymal stromal cell-derived small extracellular vesicles (MSC-sEVs) based on their ability to prevent acquisition of the M1 phenotype in lipopolysaccharide (LPS)-stimulated RAW 264.7 macrophages. Induction of tumor necrosis factor alpha, IL-1β, IL-6 and inducible nitric oxide synthase (iNOS) characterizes the M1 phenotype. Nitric oxide released by iNOS turns into nitrite, which can be easily quantitated in culture media by Griess reaction. METHODS The authors first tested different assay conditions in 96-well plates, including two seeding densities (2 × 104 cells/well and 4 × 104 cells/well), four LPS doses (1 ng/mL, 10 ng/mL, 100 ng/mL and 1000 ng/mL) and two time points (16 h and 24 h), in order to determine the best set-up to accurately measure nitrite concentration as an index of M1 macrophage polarization. RESULTS The authors found that seeding 2 × 104 cells/well and stimulating with 10 ng/mL LPS for 16 h allowed the inhibition of nitrite production by 60% with the use of dexamethasone. Using these established conditions, the authors were able to test different MSC-sEV preparations and generate dose-response curves. Moreover, the authors fully analytically validated assay performance and fulfilled cross-validation against other M1 markers. CONCLUSIONS The authors standardized a quick, cheap and reproducible in vitro macrophage assay that allows for the evaluation and estimation of the anti-inflammatory activity of MSC-sEVs.
Collapse
|
44
|
Feng ZY, Zhang QY, Tan J, Xie HQ. Techniques for increasing the yield of stem cell-derived exosomes: what factors may be involved? SCIENCE CHINA. LIFE SCIENCES 2022; 65:1325-1341. [PMID: 34637101 PMCID: PMC8506103 DOI: 10.1007/s11427-021-1997-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 08/11/2021] [Indexed: 02/05/2023]
Abstract
Exosomes are nano-scale extracellular vesicles secreted by cells and constitute an important part in the cell-cell communication. The main contents of the exosomes include proteins, microRNAs, and lipids. The mechanism and safety of stem cell-derived exosomes have rendered them a promising therapeutic strategy for regenerative medicine. Nevertheless, limited yield has restrained full explication of their functions and clinical applications To address this, various attempts have been made to explore the up- and down-stream manipulations in a bid to increase the production of exosomes. This review has recapitulated factors which may influence the yield of stem cell-derived exosomes, including selection and culture of stem cells, isolation and preservation of the exosomes, and development of artificial exosomes.
Collapse
Affiliation(s)
- Zi-Yuan Feng
- Laboratory of Stem Cell and Tissue Engineering, Orthopedic Research Institute, Med-X Center for Materials, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Qing-Yi Zhang
- Laboratory of Stem Cell and Tissue Engineering, Orthopedic Research Institute, Med-X Center for Materials, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jie Tan
- Laboratory of Stem Cell and Tissue Engineering, Orthopedic Research Institute, Med-X Center for Materials, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Hui-Qi Xie
- Laboratory of Stem Cell and Tissue Engineering, Orthopedic Research Institute, Med-X Center for Materials, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
45
|
Zhang Y, Xu C. Effects of exosomes on adult hippocampal neurogenesis and neuropsychiatric disorders. Mol Biol Rep 2022; 49:6763-6777. [PMID: 35262819 DOI: 10.1007/s11033-022-07313-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 03/01/2022] [Indexed: 12/19/2022]
Abstract
Exosomes are extracellular vesicles originating from the endosomal system, which are involved in intercellular substance transfer and cell waste elimination. Recent studies implicate the roles of exosomes in adult hippocampal neurogenesis, a process through which new granule cells are generated in the dentate gyrus, and which is closely related to mood and cognition, as well as psychiatric disorders. As such, exosomes are recognized as potential biomarkers of neurologic and psychiatric disorders. This review briefly introduces the synthesis and secretion mechanism of exosomes, and discuss the relationship between exosomes and hippocampal neurogenesis, and their roles in regulating depression, epilepsy and schizophrenia. Finally, we discuss the prospects of their application in diagnosing disorders of the central nervous system (CNS).
Collapse
Affiliation(s)
- Ying Zhang
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing, 210009, China
| | - Chi Xu
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing, 210009, China. .,Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| |
Collapse
|
46
|
Gonias SL, Banki MA, Azmoon P, Romero HK, Sigurdson CJ, Mantuano E, Campana WM. Cellular prion protein in human plasma-derived extracellular vesicles promotes neurite outgrowth via the NMDA receptor-LRP1 receptor system. J Biol Chem 2022; 298:101642. [PMID: 35090893 PMCID: PMC8861162 DOI: 10.1016/j.jbc.2022.101642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 01/08/2022] [Accepted: 01/10/2022] [Indexed: 11/22/2022] Open
Abstract
Exosomes and other extracellular vesicles (EVs) participate in cell-cell communication. Herein, we isolated EVs from human plasma and demonstrated that these EVs activate cell signaling and promote neurite outgrowth in PC-12 cells. Analysis of human plasma EVs purified by sequential ultracentrifugation using tandem mass spectrometry indicated the presence of multiple plasma proteins, including α2-macroglobulin, which is reported to regulate PC-12 cell physiology. We therefore further purified EVs by molecular exclusion or phosphatidylserine affinity chromatography, which reduced plasma protein contamination. EVs subjected to these additional purification methods exhibited unchanged activity in PC-12 cells, even though α2-macroglobulin was reduced to undetectable levels. Nonpathogenic cellular prion protein (PrPC) was carried by human plasma EVs and essential for the effects of EVs on PC-12 cells, as EV-induced cell signaling and neurite outgrowth were blocked by the PrPC-specific antibody, POM2. In addition, inhibitors of the N-methyl-d-aspartate (NMDA) receptor (NMDA-R) and low-density lipoprotein receptor-related protein-1 (LRP1) blocked the effects of plasma EVs on PC-12 cells, as did silencing of Lrp1 or the gene encoding the GluN1 NMDA-R subunit (Grin1). These results implicate the NMDA-R-LRP1 complex as the receptor system responsible for mediating the effects of EV-associated PrPC. Finally, EVs harvested from rat astrocytes carried PrPC and replicated the effects of human plasma EVs on PC-12 cell signaling. We conclude that interaction of EV-associated PrPC with the NMDA-R-LRP1 complex in target cells represents a novel mechanism by which EVs may participate in intercellular communication in the nervous system.
Collapse
Affiliation(s)
- Steven L Gonias
- Department of Pathology, University of California San Diego, La Jolla, California, USA.
| | - Michael A Banki
- Department of Pathology, University of California San Diego, La Jolla, California, USA
| | - Pardis Azmoon
- Department of Pathology, University of California San Diego, La Jolla, California, USA
| | - Haylie K Romero
- Department of Anesthesiology and Program in Neurosciences, University of California San Diego, La Jolla, California, USA
| | - Christina J Sigurdson
- Department of Pathology, University of California San Diego, La Jolla, California, USA
| | - Elisabetta Mantuano
- Department of Pathology, University of California San Diego, La Jolla, California, USA
| | - Wendy M Campana
- Department of Anesthesiology and Program in Neurosciences, University of California San Diego, La Jolla, California, USA; San Diego Veterans Administration Healthcare System, San Diego, California, USA
| |
Collapse
|
47
|
Bischoff JP, Schulz A, Morrison H. The role of exosomes in inter-cellular and inter-organ communication of the peripheral nervous system. FEBS Lett 2022; 596:655-664. [PMID: 34990014 DOI: 10.1002/1873-3468.14274] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 12/09/2021] [Accepted: 12/23/2021] [Indexed: 11/11/2022]
Abstract
Exosomes, nano-sized extracellular vesicles, are produced via the endosomal pathway and released in the extracellular space upon fusion of multivesicular bodies with the plasma membrane. Recent evidence shows that these extracellular vesicles play a key role in cell-to-cell communication. Exosomes transport bioactive proteins, messenger RNA (mRNAs) and microRNA (miRNAs) in an active form to adjacent cells or to distant organs. In this review, we focus on the role of exosomes in peripheral nerve maintenance and repair, as well as peripheral nerve/organ crosstalk, and discuss the potential benefits of exploiting exosomes for treating PNS injuries. In addition, we will highlight the emerging role of exosomes as new important vehicles for physiological systemic crosstalk failures, which could lead to organ dysfunction during neuroinflammation or aging.
Collapse
Affiliation(s)
- Julia Patricia Bischoff
- Leibniz Institute on Aging, Fritz Lipmann Institute, Beutenbergstrasse 11, 07745, Jena, Germany
| | - Alexander Schulz
- Leibniz Institute on Aging, Fritz Lipmann Institute, Beutenbergstrasse 11, 07745, Jena, Germany
| | - Helen Morrison
- Leibniz Institute on Aging, Fritz Lipmann Institute, Beutenbergstrasse 11, 07745, Jena, Germany.,Institute of Biochemistry and Biophysics, Friedrich-Schiller-University Jena, Faculty of Biological Sciences, Friedrich Schiller University Jena, Jena, Germany
| |
Collapse
|
48
|
Numerous nanoparticles as drug delivery system to control secondary immune response and promote spinal cord injury regeneration. Process Biochem 2022. [DOI: 10.1016/j.procbio.2021.11.025] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
49
|
Hercher D, Nguyen MQ, Dworak H. Extracellular vesicles and their role in peripheral nerve regeneration. Exp Neurol 2021; 350:113968. [PMID: 34973963 DOI: 10.1016/j.expneurol.2021.113968] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 12/04/2021] [Accepted: 12/25/2021] [Indexed: 12/18/2022]
Abstract
Peripheral nerve injuries often result in sensory and motor dysfunction in respective parts of the body. Regeneration after peripheral nerve injuries is a complex process including the differentiation of Schwann cells, recruiting of macrophages, blood vessel growth and axonal regrowth. Extracellular vesicles (EVs) are considered to play a pivotal role in intercellular communication and transfer of biological information. Specifically, their bioactivity and ability to deliver cargos of various types of nucleic acids and proteins have made them a potential vehicle for neurotherapeutics. However, production, characterization, dosage and targeted delivery of EVs still pose challenges for the clinical translation of EV therapeutics. This review summarizes the current knowledge of EVs in the context of the healthy and injured peripheral nerve and addresses novel concepts for modification of EVs as therapeutic agents for peripheral nerve regeneration.
Collapse
Affiliation(s)
- David Hercher
- Ludwig Boltzmann Institute for Traumatology, The Research Center in Cooperation with AUVA, Vienna, Austria; Austrian Cluster for Tissue Regeneration, Vienna, Austria.
| | - Mai Quyen Nguyen
- Ludwig Boltzmann Institute for Traumatology, The Research Center in Cooperation with AUVA, Vienna, Austria; Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Helene Dworak
- Ludwig Boltzmann Institute for Traumatology, The Research Center in Cooperation with AUVA, Vienna, Austria; Ludwig Boltzmann Research Group Senescence and Healing of Wounds, Vienna, Austria; Austrian Cluster for Tissue Regeneration, Vienna, Austria
| |
Collapse
|
50
|
Klimovich P, Rubina K, Sysoeva V, Semina E. New Frontiers in Peripheral Nerve Regeneration: Concerns and Remedies. Int J Mol Sci 2021; 22:13380. [PMID: 34948176 PMCID: PMC8703705 DOI: 10.3390/ijms222413380] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 11/30/2021] [Accepted: 12/07/2021] [Indexed: 01/08/2023] Open
Abstract
Topical advances in studying molecular and cellular mechanisms responsible for regeneration in the peripheral nervous system have highlighted the ability of the nervous system to repair itself. Still, serious injuries represent a challenge for the morphological and functional regeneration of peripheral nerves, calling for new treatment strategies that maximize nerve regeneration and recovery. This review presents the canonical view of the basic mechanisms of nerve regeneration and novel data on the role of exosomes and their transferred microRNAs in intracellular communication, regulation of axonal growth, Schwann cell migration and proliferation, and stromal cell functioning. An integrated comprehensive understanding of the current mechanistic underpinnings will open the venue for developing new clinical strategies to ensure full regeneration in the peripheral nervous system.
Collapse
Affiliation(s)
- Polina Klimovich
- National Cardiology Research Center Ministry of Health of the Russian Federation, Institute of Experimental Cardiology, 121552 Moscow, Russia; (P.K.); (E.S.)
- Faculty of Medicine, Lomonosov Moscow State University, 119991 Moscow, Russia;
| | - Kseniya Rubina
- Faculty of Medicine, Lomonosov Moscow State University, 119991 Moscow, Russia;
| | - Veronika Sysoeva
- Faculty of Medicine, Lomonosov Moscow State University, 119991 Moscow, Russia;
| | - Ekaterina Semina
- National Cardiology Research Center Ministry of Health of the Russian Federation, Institute of Experimental Cardiology, 121552 Moscow, Russia; (P.K.); (E.S.)
- Faculty of Medicine, Lomonosov Moscow State University, 119991 Moscow, Russia;
| |
Collapse
|