1
|
Bai T, Wang L, Qiao Z, Wang Z. Cuproptosis, a potential target for the therapy of diabetic critical limb ischemia. Free Radic Biol Med 2025; 234:131-140. [PMID: 40246253 DOI: 10.1016/j.freeradbiomed.2025.04.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 03/19/2025] [Accepted: 04/14/2025] [Indexed: 04/19/2025]
Abstract
Diabetic patients are considered as the high risk population to develop critical limb ischemia (CLI), a peripheral vascular disease (PVD) resulted from atherosclerosis. Cuproptosis is a novel copper-dependent cell death that has shown the regulatory role in diabetes, while its effect on diabetic CLI has not been explored yet. In this study, Diabetic CLI mice was induced by femoral artery ligation (FAL) on diabetic mice. Endothelial injury in diabetic CLI was mimicked in human microvascular endothelial cells (HMEC-1) via the induction with high glucose (HG) and nutrient deprivation (ND). Besides, copper chelator Ammonium Tetrathiomolybdate (TM), which has shown the anti-cuproptosis property, was administrated to explore its potential effects on diabetic CLI mice and HG/ND-induced HMEC-1 cells. Strikingly, obvious cuproptosis was found in the gastrocnemius muscles of diabetic CLI mice and HG/ND-induced HMEC-1 cells, as evidenced by the copper overload and dysregulated cuproptosis-related proteins (such as Fe-S cluster proteins, copper exporter ATP7A, and copper importer SLC31A1). More importantly, TM protected against the hindlimb ischemic damages in diabetic CLI mice and alleviated cuproptosis-associated cell deaths in HG/ND-induced HMEC-1 cells. In summary, this study indicates the involvements of cuproptosis in diabetic CLI, and provides novel insights into copper chelator TM on diabetic CLI therapy.
Collapse
Affiliation(s)
- Tao Bai
- Department of Vascular and Endovascular Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
| | - Luhao Wang
- Department of Vascular and Endovascular Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Zhentao Qiao
- Department of Vascular and Endovascular Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Zhiwei Wang
- Department of Vascular and Endovascular Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
| |
Collapse
|
2
|
Che Y, Shimizu Y, Murohara T. Therapeutic Potential of Adipose-Derived Regenerative Cells for Ischemic Diseases. Cells 2025; 14:343. [PMID: 40072072 PMCID: PMC11898683 DOI: 10.3390/cells14050343] [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: 01/22/2025] [Revised: 02/21/2025] [Accepted: 02/26/2025] [Indexed: 03/15/2025] Open
Abstract
Adipose-derived regenerative cells (ADRCs) are one of the most promising cell sources that possess significant therapeutic effects. They have now become a main source of cell therapy for the treatment of ischemic diseases due to their easy accessibility, expansion, and differentiation. Additionally, ADRCs can release multiple paracrine factors and extracellular vesicles that contribute to tissue regeneration by promoting angiogenesis, regulating inflammation, alleviating apoptosis, and inhibiting fibrosis. However, ADRCs still have some limitations to realize their full therapeutic potential. To address these issues, protective mechanistic studies and bioengineering studies have been carried out. This review focused on the recently studied mechanisms, such as paracrine factors, cell fusion, and mitochondrial transfer, involving the therapeutic potential of ADRCs in ischemic diseases and discussed some modification techniques of ADRCs.
Collapse
Affiliation(s)
| | - Yuuki Shimizu
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | | |
Collapse
|
3
|
Rosina M, Scaricamazza S, Riggio F, Fenili G, Giannessi F, Matteocci A, Nesci V, Salvatori I, Angelini DF, Aquilano K, Chiurchiù V, Barbato DL, Mercuri NB, Valle C, Ferri A. Brown Adipose Tissue undergoes pathological perturbations and shapes C2C12 myoblast homeostasis in the SOD1-G93A mouse model of Amyotrophic Lateral Sclerosis. Heliyon 2025; 11:e41801. [PMID: 39916853 PMCID: PMC11800085 DOI: 10.1016/j.heliyon.2025.e41801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 12/04/2024] [Accepted: 01/07/2025] [Indexed: 02/09/2025] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a progressive neurodegenerative disease characterized by the selective loss of motor neurons. The contribution of peripheral organs remains incompletely understood. We focused our attention on brown adipose tissue (BAT) and its secreted extracellular vesicles (EVs) given their role in regulating systemic energy balance. In this study, we employed a multi-omics approach, including RNA sequencing (GEO identifier GSE273052) and proteomics (ProteomeXchange identifier PXD054147), to investigate the alterations in BAT and its EVs in the SOD1-G93A mouse model of ALS. Our results revealed consistent changes in the proteomic and transcriptomic profiles of BAT from SOD1-G93A mice, highlighting alterations such as mitochondrial dysfunction and impaired differentiation capacity. Specifically, primary brown adipocytes (PBAs) from SOD1-G93A mice exhibited differentiation impairment, respiratory defects, and alterations in mitochondrial dynamics. Furthermore, the BAT-derived EVs from SOD1-G93A mice displayed distinct changes in size distribution and cargo content. In parallel, such EVs negatively impacted the differentiation and homeostasis of C2C12 murine myoblasts, as well as induced atrophy in C2C12-derived myotubes. These findings suggest that BAT undergoes pathological perturbations in ALS mouse model and could impact on skeletal muscle homeostasis through the secretion of dysfunctional EVs.
Collapse
Affiliation(s)
- Marco Rosina
- Unit of Neurology, Fondazione PTV Policlinico Tor Vergata, Viale Oxford 81, 00133, Rome, Italy
- Laboratory of Neurochemistry, IRCCS Fondazione Santa Lucia, Via del fosso di fiorano 64, 00143, Rome, Italy
| | - Silvia Scaricamazza
- Laboratory of Neurochemistry, IRCCS Fondazione Santa Lucia, Via del fosso di fiorano 64, 00143, Rome, Italy
- Institute of Translational Pharmacology, National Research Council, Via del fosso del cavaliere 100, 00133, Rome, Italy
| | - Flaminia Riggio
- Department of Biology and Biotechnology “Charles Darwin”, University of Roma “La Sapienza”, 00161, Rome, Italy
| | - Gianmarco Fenili
- Laboratory of Neurochemistry, IRCCS Fondazione Santa Lucia, Via del fosso di fiorano 64, 00143, Rome, Italy
- Department of Movement, Human and Health Sciences University of Rome "Foro Italico”, Piazza Lauro de Bosis 6, 00135, Rome, Italy
| | - Flavia Giannessi
- Laboratory of Molecular Virology and Antimicrobial Immunity, Department of Science, Roma Tre University, 00146, Rome, Italy
- Neuroimmunology Unit, IRCCS Fondazione Santa Lucia, Via del fosso di fiorano 64, 00143, Rome, Italy
| | - Alessandro Matteocci
- Laboratory of Resolution of Neuroinflammation, IRCCS Fondazione Santa Lucia, Via del fosso di fiorano 64, 00143, Rome, Italy
- PhD program in Immunology, Molecular Medicine and Applied biotechnologies, University of Rome “Tor Vergata”, 00133, Rome, Italy
| | - Valentina Nesci
- Laboratory of Neurochemistry, IRCCS Fondazione Santa Lucia, Via del fosso di fiorano 64, 00143, Rome, Italy
- Department of Systems Medicine, University of Roma "Tor Vergata", 00133, Rome, Italy
| | - Illari Salvatori
- Laboratory of Neurochemistry, IRCCS Fondazione Santa Lucia, Via del fosso di fiorano 64, 00143, Rome, Italy
- Department of Experimental Medicine, University of Roma "La Sapienza", 00161, Rome, Italy
| | - Daniela F. Angelini
- Neuroimmunology Unit, IRCCS Fondazione Santa Lucia, Via del fosso di fiorano 64, 00143, Rome, Italy
| | - Katia Aquilano
- Department of Biology, University of Rome “Tor Vergata”, via della ricerca scientifica, 00133, Rome, Italy
| | - Valerio Chiurchiù
- Institute of Translational Pharmacology, National Research Council, Via del fosso del cavaliere 100, 00133, Rome, Italy
- Laboratory of Resolution of Neuroinflammation, IRCCS Fondazione Santa Lucia, Via del fosso di fiorano 64, 00143, Rome, Italy
| | - Daniele Lettieri Barbato
- Department of Biology, University of Rome “Tor Vergata”, via della ricerca scientifica, 00133, Rome, Italy
| | - Nicola Biagio Mercuri
- Unit of Neurology, Fondazione PTV Policlinico Tor Vergata, Viale Oxford 81, 00133, Rome, Italy
- Department of Systems Medicine, University of Roma "Tor Vergata", 00133, Rome, Italy
- Laboratory of Experimental Neurology, IRCCS Fondazione Santa Lucia, Via del fosso di fiorano 64, 00143, Rome, Italy
| | - Cristiana Valle
- Laboratory of Neurochemistry, IRCCS Fondazione Santa Lucia, Via del fosso di fiorano 64, 00143, Rome, Italy
- Institute of Translational Pharmacology, National Research Council, Via del fosso del cavaliere 100, 00133, Rome, Italy
| | - Alberto Ferri
- Laboratory of Neurochemistry, IRCCS Fondazione Santa Lucia, Via del fosso di fiorano 64, 00143, Rome, Italy
- Institute of Translational Pharmacology, National Research Council, Via del fosso del cavaliere 100, 00133, Rome, Italy
| |
Collapse
|
4
|
Ravi Mythili VM, Rajendran RL, Arun R, Thasma Loganathbabu VK, Reyaz D, Nagarajan AK, Ahn BC, Gangadaran P. Emerging Strategies for Revascularization: Use of Cell-Derived Extracellular Vesicles and Artificial Nanovesicles in Critical Limb Ischemia. Bioengineering (Basel) 2025; 12:92. [PMID: 39851366 PMCID: PMC11762151 DOI: 10.3390/bioengineering12010092] [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: 11/27/2024] [Revised: 01/09/2025] [Accepted: 01/15/2025] [Indexed: 01/26/2025] Open
Abstract
Critical limb ischemia (CLI) poses a substantial and intricate challenge in vascular medicine, necessitating the development of innovative therapeutic strategies to address its multifaceted pathophysiology. Conventional revascularization approaches often fail to adequately address the complexity of CLI, necessitating the identification of alternative methodologies. This review explores uncharted territory beyond traditional therapies, focusing on the potential of two distinct yet interrelated entities: cell-derived extracellular vesicles (EVs) and artificial nanovesicles. Cell-derived EVs are small membranous structures naturally released by cells, and artificial nanovesicles are artificially engineered nanosized vesicles. Both these vesicles represent promising avenues for therapeutic intervention. They act as carriers of bioactive cargo, including proteins, nucleic acids, and lipids, that can modulate intricate cellular responses associated with ischemic tissue repair and angiogenesis. This review also assesses the evolving landscape of CLI revascularization through the unique perspective of cell-derived EVs and artificial nanovesicles. The review spans the spectrum from early preclinical investigations to the latest translational advancements, providing a comprehensive overview of the current state of research in this emerging field. These groundbreaking vesicle therapies hold immense potential for revolutionizing CLI treatment paradigms.
Collapse
Affiliation(s)
- Vijay Murali Ravi Mythili
- Integrative Genetics and Molecular Oncology Group, Department of Genetic Engineering, College of Engineering and Technology, SRM Institute of Science and Technology, Chengalpattu 603203, Tamil Nadu, India; (V.M.R.M.); (R.A.); (V.K.T.L.); (D.R.); (A.K.N.)
| | - Ramya Lakshmi Rajendran
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea;
- BK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Department of Biomedical Sciences, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
- Cardiovascular Research Institute, Kyungpook National University, Daegu 41944, Republic of Korea
| | - Raksa Arun
- Integrative Genetics and Molecular Oncology Group, Department of Genetic Engineering, College of Engineering and Technology, SRM Institute of Science and Technology, Chengalpattu 603203, Tamil Nadu, India; (V.M.R.M.); (R.A.); (V.K.T.L.); (D.R.); (A.K.N.)
| | - Vasanth Kanth Thasma Loganathbabu
- Integrative Genetics and Molecular Oncology Group, Department of Genetic Engineering, College of Engineering and Technology, SRM Institute of Science and Technology, Chengalpattu 603203, Tamil Nadu, India; (V.M.R.M.); (R.A.); (V.K.T.L.); (D.R.); (A.K.N.)
| | - Danyal Reyaz
- Integrative Genetics and Molecular Oncology Group, Department of Genetic Engineering, College of Engineering and Technology, SRM Institute of Science and Technology, Chengalpattu 603203, Tamil Nadu, India; (V.M.R.M.); (R.A.); (V.K.T.L.); (D.R.); (A.K.N.)
| | - ArulJothi Kandasamy Nagarajan
- Integrative Genetics and Molecular Oncology Group, Department of Genetic Engineering, College of Engineering and Technology, SRM Institute of Science and Technology, Chengalpattu 603203, Tamil Nadu, India; (V.M.R.M.); (R.A.); (V.K.T.L.); (D.R.); (A.K.N.)
| | - Byeong-Cheol Ahn
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea;
- BK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Department of Biomedical Sciences, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
- Cardiovascular Research Institute, Kyungpook National University, Daegu 41944, Republic of Korea
- Department of Nuclear Medicine, Kyungpook National University Hospital, Daegu 41944, Republic of Korea
| | - Prakash Gangadaran
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea;
- BK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Department of Biomedical Sciences, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
- Cardiovascular Research Institute, Kyungpook National University, Daegu 41944, Republic of Korea
| |
Collapse
|
5
|
Wang T, Zhou D, Hong Z. Sarcopenia and cachexia: molecular mechanisms and therapeutic interventions. MedComm (Beijing) 2025; 6:e70030. [PMID: 39764565 PMCID: PMC11702502 DOI: 10.1002/mco2.70030] [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: 07/16/2024] [Revised: 11/11/2024] [Accepted: 11/12/2024] [Indexed: 03/17/2025] Open
Abstract
Sarcopenia is defined as a muscle-wasting syndrome that occurs with accelerated aging, while cachexia is a severe wasting syndrome associated with conditions such as cancer and immunodeficiency disorders, which cannot be fully addressed through conventional nutritional supplementation. Sarcopenia can be considered a component of cachexia, with the bidirectional interplay between adipose tissue and skeletal muscle potentially serving as a molecular mechanism for both conditions. However, the underlying mechanisms differ. Recognizing the interplay and distinctions between these disorders is essential for advancing both basic and translational research in this area, enhancing diagnostic accuracy and ultimately achieving effective therapeutic solutions for affected patients. This review discusses the muscle microenvironment's changes contributing to these conditions, recent therapeutic approaches like lifestyle modifications, small molecules, and nutritional interventions, and emerging strategies such as gene editing, stem cell therapy, and gut microbiome modulation. We also address the challenges and opportunities of multimodal interventions, aiming to provide insights into the pathogenesis and molecular mechanisms of sarcopenia and cachexia, ultimately aiding in innovative strategy development and improved treatments.
Collapse
Affiliation(s)
- Tiantian Wang
- Department of NeurologyWest China Hospital of Sichuan UniversityChengduSichuanChina
- Institute of Brain Science and Brain‐Inspired Technology of West China HospitalSichuan UniversityChengduSichuanChina
- Department of NeurologyChengdu Shangjin Nanfu HospitalChengduSichuanChina
| | - Dong Zhou
- Department of NeurologyWest China Hospital of Sichuan UniversityChengduSichuanChina
- Institute of Brain Science and Brain‐Inspired Technology of West China HospitalSichuan UniversityChengduSichuanChina
- Department of NeurologyChengdu Shangjin Nanfu HospitalChengduSichuanChina
| | - Zhen Hong
- Department of NeurologyWest China Hospital of Sichuan UniversityChengduSichuanChina
- Institute of Brain Science and Brain‐Inspired Technology of West China HospitalSichuan UniversityChengduSichuanChina
- Department of NeurologyChengdu Shangjin Nanfu HospitalChengduSichuanChina
| |
Collapse
|
6
|
Sun X, Ding H, Li X, Wu Y, Huang X. Disulfiram-loaded nanovesicles hydrogel promotes healing of diabetic wound. J Transl Med 2024; 22:1066. [PMID: 39593097 PMCID: PMC11600750 DOI: 10.1186/s12967-024-05875-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Accepted: 11/11/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND Traditional methods for treating diabetic wounds are limited in effectiveness because of their long healing times, the risk of immune rejection, and susceptibility to infection. Suppressing neutrophil extracellular traps (NETs) is an effective strategy for reducing persistent inflammation in diabetic wounds. Although disulfiram (DSF) can inhibit the significant increase of NETs in diabetic wounds, oral DSF suffers from rapid and harmful metabolism in the liver. To address these challenges, we developed a nanomedicine formulation in which DSF was incorporated into the hydrogel. METHODS In this study, we developed a DSF-laden sodium alginate hydrogel wound dressing, DEP@SA, and characterized its composition, properties, and performance. We examined the effects of DEP@SA on inflammatory phase-related markers such as NETs and their pathway proteins, inflammatory factors, and macrophage phenotypes in a high-glucose environment in vivo and in vitro. In addition, the effects of DEP@SA on tissue regenerative capacity such as epidermal proliferative migration and angiogenesis, were also assessed. RESULTS The results showed that by utilizing extracellular vesicles as a drug delivery system, we effectively mitigated the degradation of DSF via direct contact with aqueous solutions and ensured the stability of DSF@SA, which could then be applied to diabetic wounds. The inflammatory phase-related indicators revealed that DSF@SA effectively reduced inflammation levels, decreased NETs formation, suppressed the Caspase-1/GSDMD pathway in neutrophils, and promoted the polarization of M2 macrophages. Moreover, the hydrogel accelerated wound healing by promoting angiogenesis and re-epithelialization, thereby shortening the diabetic wound healing time. CONCLUSIONS This study confirmed that the DSF@SA composite dressing has the potential to enhance diabetic wound repair and offers a novel approach for drug reutilization.
Collapse
Affiliation(s)
- Xingzi Sun
- Center for Infection and Immunity and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, 519000, China
| | - Hanxi Ding
- Department of Oncology, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, 519000, China
| | - Xingyu Li
- Center for Infection and Immunity and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, 519000, China
| | - Yongjian Wu
- Center for Infection and Immunity and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, 519000, China.
| | - Xi Huang
- Center for Infection and Immunity and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, 519000, China.
| |
Collapse
|
7
|
Rahnama M, Heidari M, Poursalehi Z, Golchin A. Global Trends of Exosomes Application in Clinical Trials: A Scoping Review. Stem Cell Rev Rep 2024; 20:2165-2193. [PMID: 39340738 DOI: 10.1007/s12015-024-10791-7] [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: 09/19/2024] [Indexed: 09/30/2024]
Abstract
BACKGROUND Exosomes, nano-sized extracellular vesicles, have emerged as a promising tool for the diagnosis and treatment of various intractable diseases, including chronic wounds and cancers. As our understanding of exosomes continues to grow, their potential as a powerful therapeutic modality in medicine is also expanding. This systematic review aims to examine the progress of exosome-based clinical trials and provide a comprehensive overview of the therapeutic perspectives of exosomes. METHODS This systematic review strictly follows PRISMA guidelines and has been registered in PROSPERO, the International Prospective Register of Systematic Reviews. It encompasses articles from January 2000 to January 2023, sourced from bibliographic databases, with targeted search terms targeting exosome applications in clinical trials. During the screening process, strict inclusion and exclusion criteria were applied, including a focus on clinical trials utilizing different cell-derived exosomes for therapeutic purposes. RESULTS Among the 522 publications initially identified, only 10 studies met the stringent eligibility criteria after meticulous screening. The selection process involved systematically excluding duplicates and irrelevant articles to provide a transparent overview. CONCLUSION According to our systematic review, exosomes have promising applications in a variety of medical fields, including cell-free therapies and drug delivery systems for treating a variety of diseases, especially cancers and chronic wounds. To ensure safety, potency, and broader clinical applications, further optimization of exosome extraction, loading, targeting, and administration is necessary. While cell-based therapeutics are increasingly utilizing exosomes, this field is still in its infancy, and ongoing clinical trials will provide valuable insights into the clinical utility of exosomes.
Collapse
Affiliation(s)
- Maryam Rahnama
- Student Research Committee, Urmia University of Medical Sciences, Urmia, Iran
- Department of Applied Cell Sciences, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Mohammad Heidari
- Department of Biostatistics and Epidemiology, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Zahra Poursalehi
- Student Research Committee, Urmia University of Medical Sciences, Urmia, Iran
- Department of Applied Cell Sciences, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Ali Golchin
- Student Research Committee, Urmia University of Medical Sciences, Urmia, Iran.
- Department of Applied Cell Sciences, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran.
| |
Collapse
|
8
|
Wang Y, Tan PC, Xu X, Zhou S. Protective function of adipocyte-derived extracellular vesicles and adipose stem cells in damage repair and regeneration. CHINESE JOURNAL OF PLASTIC AND RECONSTRUCTIVE SURGERY 2024. [DOI: 10.1016/j.cjprs.2024.11.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
9
|
Zhang Y, Wu D, Zhou C, Bai M, Wan Y, Zheng Q, Fan Z, Wang X, Yang C. Engineered extracellular vesicles for tissue repair and regeneration. BURNS & TRAUMA 2024; 12:tkae062. [PMID: 39439545 PMCID: PMC11495891 DOI: 10.1093/burnst/tkae062] [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: 07/09/2024] [Revised: 09/12/2024] [Accepted: 09/21/2024] [Indexed: 10/25/2024]
Abstract
Extracellular vesicles (EVs) are heterogeneous membrane-like vesicles secreted by living cells that are involved in many physiological and pathological processes and act as intermediaries of intercellular communication and molecular transfer. Recent studies have shown that EVs from specific sources regulate tissue repair and regeneration by delivering proteins, lipids, and nucleic acids to target cells as signaling molecules. Nanotechnology breakthroughs have facilitated the development and exploration of engineered EVs for tissue repair. Enhancements through gene editing, surface modification, and content modification have further improved their therapeutic efficacy. This review summarizes the potential of EVs in tissue repair and regeneration, their mechanisms of action, and their research progress in regenerative medicine. This review highlights their design logic through typical examples and explores the development prospects of EVs in tissue repair. The aim of this review is to provide new insights into the design of EVs for tissue repair and regeneration applications, thereby expanding their use in regenerative medicine.
Collapse
Affiliation(s)
- Yan Zhang
- College of Basic Medicin, Beihua University, No. 3999 Binjiang East Road, Fengman District, Jilin City, Jilin Province, China
- School of Public Health, Beihua University, No. 3999 Binjiang East Road, Fengman District, Jilin City, Jilin Province, China
| | - Dan Wu
- College of Basic Medicin, Beihua University, No. 3999 Binjiang East Road, Fengman District, Jilin City, Jilin Province, China
| | - Chen Zhou
- Department of Laboratory Medicine, The Eighth Affiliated Hospital, Sun Yat-Sen University, No. 3025 Shennan Middle Road, Futian District, Shenzhen, China
| | - Muran Bai
- College of Basic Medicin, Beihua University, No. 3999 Binjiang East Road, Fengman District, Jilin City, Jilin Province, China
| | - Yucheng Wan
- Hospital of Stomatology, Zunyi Medical University, No. 89, Wujiang East Road, Xinpu New District, Zunyi City, Guizhou Province, China
| | - Qing Zheng
- College of Basic Medicin, Beihua University, No. 3999 Binjiang East Road, Fengman District, Jilin City, Jilin Province, China
| | - Zhijin Fan
- Institute for Engineering Medicine, Kunming Medical University, No. 1168 Chunrong West Road, Yuhua Street, Chenggong District, Kunming City, Yunnan Province China
| | - Xianwen Wang
- School of Biomedical Engineering, Research and Engineering Center of Biomedical Materials, Anhui Medical University, No.81 Meishan Road, Shushan District, Hefei 230032, China
| | - Chun Yang
- College of Basic Medicin, Beihua University, No. 3999 Binjiang East Road, Fengman District, Jilin City, Jilin Province, China
| |
Collapse
|
10
|
Liu S, Zhao H, Jiang T, Wan G, Yan C, Zhang C, Yang X, Chen Z. The Angiogenic Repertoire of Stem Cell Extracellular Vesicles: Demystifying the Molecular Underpinnings for Wound Healing Applications. Stem Cell Rev Rep 2024; 20:1795-1812. [PMID: 39001965 DOI: 10.1007/s12015-024-10762-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/06/2024] [Indexed: 07/15/2024]
Abstract
Stem cells-derived extracellular vesicles (SC-EVs) have emerged as promising therapeutic agents for wound repair, recapitulating the biological effects of parent cells while mitigating immunogenic and tumorigenic risks. These EVs orchestrate wound healing processes, notably through modulating angiogenesis-a critical event in tissue revascularization and regeneration. This study provides a comprehensive overview of the multifaceted mechanisms underpinning the pro-angiogenic capacity of EVs from various stem cell sources within the wound microenvironment. By elucidating the molecular intricacies governing their angiogenic prowess, we aim to unravel the mechanistic repertoire underlying their remarkable potential to accelerate wound healing. Additionally, methods to enhance the angiogenic effects of SC-EVs, current limitations, and future perspectives are highlighted, emphasizing the significant potential of this rapidly advancing field in revolutionizing wound healing strategies.
Collapse
Affiliation(s)
- Shuoyuan Liu
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Huayuan Zhao
- Department of Urology, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Tao Jiang
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Gui Wan
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Chengqi Yan
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Chi Zhang
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xiaofan Yang
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Zhenbing Chen
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
11
|
Zhu X, Yu G, Lv Y, Yang N, Zhao Y, Li F, Zhao J, Chen Z, Lai Y, Chen L, Wang X, Xiao J, Cai Y, Feng Y, Ding J, Gao W, Zhou K, Xu H. Neuregulin-1, a member of the epidermal growth factor family, mitigates STING-mediated pyroptosis and necroptosis in ischaemic flaps. BURNS & TRAUMA 2024; 12:tkae035. [PMID: 38855574 PMCID: PMC11162832 DOI: 10.1093/burnst/tkae035] [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: 11/15/2023] [Revised: 05/22/2024] [Accepted: 05/22/2024] [Indexed: 06/11/2024]
Abstract
Background Ensuring the survival of the distal end of a random flap during hypoperfusion (ischaemia) is difficult in clinical practice. Effective prevention of programmed cell death is a potential strategy for inhibiting ischaemic flap necrosis. The activation of stimulator of interferon genes (STING) pathway promotes inflammation and leads to cell death. The epidermal growth factor family member neuregulin-1 (NRG1) reduces cell death by activating the protein kinase B (AKT) signalling pathway. Moreover, AKT signalling negatively regulates STING activity. We aimed to verify the efficacy of NRG1 injection in protecting against flap necrosis. Additionally, we investigated whether NRG1 effectively enhances ischemic flap survival by inhibiting pyroptosis and necroptosis through STING suppression. Methods A random-pattern skin flap model was generated on the backs of C57BL/6 mice. The skin flap survival area was determined. The blood supply and vascular network of the flap was assessed by laser Doppler blood flow analysis. Cluster of differentiation 34 immunohistochemistry (IHC) and haematoxylin and eosin (H&E) staining of the flap sections revealed microvessels. Transcriptome sequencing analysis revealed the mechanism by which NRG1 promotes the survival of ischaemic flaps. The levels of angiogenesis, oxidative stress, necroptosis, pyroptosis and indicators associated with signalling pathways in flaps were examined by IHC, immunofluorescence and Western blotting. Packaging adeno-associated virus (AAV) was used to activate STING in flaps. Results NRG1 promoted the survival of ischaemic flaps. An increased subcutaneous vascular network and neovascularization were found in ischaemic flaps after the application of NRG1. Transcriptomic gene ontology enrichment analysis and protein level detection indicated that necroptosis, pyroptosis and STING activity were reduced in the NRG1 group. The phosphorylation of AKT and forkhead box O3a (FOXO3a) were increased after NRG1 treatment. The increased expression of STING in flaps induced by AAV reversed the therapeutic effect of NRG1. The ability of NRG1 to phosphorylate AKT-FOXO3a, inhibit STING and promote flap survival was abolished after the application of the AKT inhibitor MK2206. Conclusions NRG1 inhibits pyroptosis and necroptosis by activating the AKT-FOXO3a signalling pathway to suppress STING activation and promote ischaemic flap survival.
Collapse
Affiliation(s)
- Xuwei Zhu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, No. 109 West Xueyuan Road, Lucheng District, Wenzhou 325027, China
- Zhejiang Provincial Key Laboratory of Orthopaedics, No. 109 West Xueyuan Road, Lucheng District, Wenzhou 325027, China
- The Second Clinical Medical College of Wenzhou Medical University, No. 109 West Xueyuan Road, Lucheng District, Wenzhou 325027, China
| | - Gaoxiang Yu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, No. 109 West Xueyuan Road, Lucheng District, Wenzhou 325027, China
- Zhejiang Provincial Key Laboratory of Orthopaedics, No. 109 West Xueyuan Road, Lucheng District, Wenzhou 325027, China
- The Second Clinical Medical College of Wenzhou Medical University, No. 109 West Xueyuan Road, Lucheng District, Wenzhou 325027, China
| | - Ya Lv
- The First Affiliated Hospital of Wenzhou Medical University, Nanbaixiang Street, Ouhai District, Wenzhou 325000, China
| | - Ningning Yang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, No. 109 West Xueyuan Road, Lucheng District, Wenzhou 325027, China
- Zhejiang Provincial Key Laboratory of Orthopaedics, No. 109 West Xueyuan Road, Lucheng District, Wenzhou 325027, China
- The Second Clinical Medical College of Wenzhou Medical University, No. 109 West Xueyuan Road, Lucheng District, Wenzhou 325027, China
| | - Yinuo Zhao
- School of Pharmaceutical Science of Zhejiang Chinese Medical University, NO. 548 Binwen Road, Binjiang District, Hangzhou 310000, China
| | - Feida Li
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, No. 109 West Xueyuan Road, Lucheng District, Wenzhou 325027, China
- Zhejiang Provincial Key Laboratory of Orthopaedics, No. 109 West Xueyuan Road, Lucheng District, Wenzhou 325027, China
- The Second Clinical Medical College of Wenzhou Medical University, No. 109 West Xueyuan Road, Lucheng District, Wenzhou 325027, China
| | - Jiayi Zhao
- The Second Clinical Medical College of Wenzhou Medical University, No. 109 West Xueyuan Road, Lucheng District, Wenzhou 325027, China
| | - Zhuliu Chen
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, No. 109 West Xueyuan Road, Lucheng District, Wenzhou 325027, China
- Zhejiang Provincial Key Laboratory of Orthopaedics, No. 109 West Xueyuan Road, Lucheng District, Wenzhou 325027, China
- The Second Clinical Medical College of Wenzhou Medical University, No. 109 West Xueyuan Road, Lucheng District, Wenzhou 325027, China
| | - Yingying Lai
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, No. 109 West Xueyuan Road, Lucheng District, Wenzhou 325027, China
- Zhejiang Provincial Key Laboratory of Orthopaedics, No. 109 West Xueyuan Road, Lucheng District, Wenzhou 325027, China
- The Second Clinical Medical College of Wenzhou Medical University, No. 109 West Xueyuan Road, Lucheng District, Wenzhou 325027, China
| | - Liang Chen
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, No. 109 West Xueyuan Road, Lucheng District, Wenzhou 325027, China
- Zhejiang Provincial Key Laboratory of Orthopaedics, No. 109 West Xueyuan Road, Lucheng District, Wenzhou 325027, China
- The Second Clinical Medical College of Wenzhou Medical University, No. 109 West Xueyuan Road, Lucheng District, Wenzhou 325027, China
| | - Xiangyang Wang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, No. 109 West Xueyuan Road, Lucheng District, Wenzhou 325027, China
- Zhejiang Provincial Key Laboratory of Orthopaedics, No. 109 West Xueyuan Road, Lucheng District, Wenzhou 325027, China
- The Second Clinical Medical College of Wenzhou Medical University, No. 109 West Xueyuan Road, Lucheng District, Wenzhou 325027, China
| | - Jian Xiao
- Molecular Pharmacology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Chashan University Town, Ouhai District, Wenzhou, 325000, China
| | - Yuepiao Cai
- Molecular Pharmacology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Chashan University Town, Ouhai District, Wenzhou, 325000, China
| | | | - Jian Ding
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, No. 109 West Xueyuan Road, Lucheng District, Wenzhou 325027, China
- Zhejiang Provincial Key Laboratory of Orthopaedics, No. 109 West Xueyuan Road, Lucheng District, Wenzhou 325027, China
- The Second Clinical Medical College of Wenzhou Medical University, No. 109 West Xueyuan Road, Lucheng District, Wenzhou 325027, China
| | - Weiyang Gao
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, No. 109 West Xueyuan Road, Lucheng District, Wenzhou 325027, China
- Zhejiang Provincial Key Laboratory of Orthopaedics, No. 109 West Xueyuan Road, Lucheng District, Wenzhou 325027, China
- The Second Clinical Medical College of Wenzhou Medical University, No. 109 West Xueyuan Road, Lucheng District, Wenzhou 325027, China
| | - Kailiang Zhou
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, No. 109 West Xueyuan Road, Lucheng District, Wenzhou 325027, China
- Zhejiang Provincial Key Laboratory of Orthopaedics, No. 109 West Xueyuan Road, Lucheng District, Wenzhou 325027, China
- The Second Clinical Medical College of Wenzhou Medical University, No. 109 West Xueyuan Road, Lucheng District, Wenzhou 325027, China
| | - Hui Xu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, No. 109 West Xueyuan Road, Lucheng District, Wenzhou 325027, China
- Zhejiang Provincial Key Laboratory of Orthopaedics, No. 109 West Xueyuan Road, Lucheng District, Wenzhou 325027, China
- The Second Clinical Medical College of Wenzhou Medical University, No. 109 West Xueyuan Road, Lucheng District, Wenzhou 325027, China
| |
Collapse
|
12
|
Yang S, Sun Y, Yan C. Recent advances in the use of extracellular vesicles from adipose-derived stem cells for regenerative medical therapeutics. J Nanobiotechnology 2024; 22:316. [PMID: 38844939 PMCID: PMC11157933 DOI: 10.1186/s12951-024-02603-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 05/28/2024] [Indexed: 06/09/2024] Open
Abstract
Adipose-derived stem cells (ADSCs) are a subset of mesenchymal stem cells (MSCs) isolated from adipose tissue. They possess remarkable properties, including multipotency, self-renewal, and easy clinical availability. ADSCs are also capable of promoting tissue regeneration through the secretion of various cytokines, factors, and extracellular vesicles (EVs). ADSC-derived EVs (ADSC-EVs) act as intercellular signaling mediators that encapsulate a range of biomolecules. These EVs have been found to mediate the therapeutic activities of donor cells by promoting the proliferation and migration of effector cells, facilitating angiogenesis, modulating immunity, and performing other specific functions in different tissues. Compared to the donor cells themselves, ADSC-EVs offer advantages such as fewer safety concerns and more convenient transportation and storage for clinical application. As a result, these EVs have received significant attention as cell-free therapeutic agents with potential future application in regenerative medicine. In this review, we focus on recent research progress regarding regenerative medical use of ADSC-EVs across various medical conditions, including wound healing, chronic limb ischemia, angiogenesis, myocardial infarction, diabetic nephropathy, fat graft survival, bone regeneration, cartilage regeneration, tendinopathy and tendon healing, peripheral nerve regeneration, and acute lung injury, among others. We also discuss the underlying mechanisms responsible for inducing these therapeutic effects. We believe that deciphering the biological properties, therapeutic effects, and underlying mechanisms associated with ADSC-EVs will provide a foundation for developing a novel therapeutic approach in regenerative medicine.
Collapse
Affiliation(s)
- Song Yang
- Institute of Life Sciences, Chongqing Medical University, Chongqing, 400016, People's Republic of China.
| | - Yiran Sun
- School of Pharmacy, Chengdu Medical College, Chengdu, 610500, People's Republic of China.
| | - Chenchen Yan
- School of Pharmacy, Chengdu Medical College, Chengdu, 610500, People's Republic of China
| |
Collapse
|
13
|
Che D, Xiang X, Xie J, Chen Z, Bao Q, Cao D. Exosomes Derived from Adipose Stem Cells Enhance Angiogenesis in Diabetic Wound Via miR-146a-5p/JAZF1 Axis. Stem Cell Rev Rep 2024; 20:1026-1039. [PMID: 38393667 PMCID: PMC11087353 DOI: 10.1007/s12015-024-10685-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/24/2024] [Indexed: 02/25/2024]
Abstract
Chronic trauma in diabetes is a leading cause of disability and mortality. Exosomes show promise in tissue regeneration. This study investigates the role of exosomes derived from adipose stem cells (ADSC-Exos) in angiogenesis. MiRNA-seq analysis revealed significant changes in 47 genes in human umbilical vein endothelial cells (HUVECs) treated with ADSC-Exos, with miR-146a-5p highly expressed. MiR-146a-5p mimics enhanced the pro-angiogenic effects of ADSC-Exos, while inhibitors had the opposite effect. JAZF1 was identified as a direct downstream target of miR-146a-5p through bioinformatics, qRT-PCR, and dual luciferase assay. Overexpress of JAZF1 resulted in decreased proliferation, migration, and angiogenic capacity of HUVECs, and reduced VEGFA expression. This study proposes that ADSC-Exos regulate angiogenesis partly via the miR-146a-5p/JAZF1 axis.
Collapse
Affiliation(s)
- Dehui Che
- Department of Plastic and Reconstructive, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Xinjian Xiang
- Department of Plastic and Reconstructive, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Juan Xie
- Department of Plastic and Reconstructive, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Zenghong Chen
- Department of Plastic and Reconstructive, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Qiong Bao
- Department of Plastic and Reconstructive, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Dongsheng Cao
- Department of Plastic and Reconstructive, The Second Affiliated Hospital of Anhui Medical University, Hefei, China.
| |
Collapse
|
14
|
Zou A, Xiao T, Chi B, Wang Y, Mao L, Cai D, Gu Q, Chen Q, Wang Q, Ji Y, Sun L. Engineered Exosomes with Growth Differentiation Factor-15 Overexpression Enhance Cardiac Repair After Myocardial Injury. Int J Nanomedicine 2024; 19:3295-3314. [PMID: 38606373 PMCID: PMC11007405 DOI: 10.2147/ijn.s454277] [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: 12/10/2023] [Accepted: 03/25/2024] [Indexed: 04/13/2024] Open
Abstract
Background Cardiac repair remains a thorny issue for survivors of acute myocardial infarction (AMI), due to the regenerative inertia of myocardial cells. Cell-free therapies, such as exosome transplantation, have become a potential strategy for myocardial injury. The aim of this study was to investigate the role of engineered exosomes in overexpressing Growth Differentiation Factor-15 (GDF-15) (GDF15-EVs) after myocardial injury, and their molecular mechanisms in cardiac repair. Methods H9C2 cells were transfected with GDF-15 lentivirus or negative control. The exosomes secreted from H9C2 cells were collected and identified. The cellular apoptosis and autophagy of H2O2-injured H9C2 cells were assessed by Western blotting, TUNEL assay, electron microscopy, CCK-8 and caspase 3/7 assay. A rat model of AMI was constructed by ligating the left anterior descending artery. The anti-apoptotic, pro-angiogenic effects of GDF15-EVs treatment, as well as ensuing functional and histological recovery were evaluated. Then, mRNA sequencing was performed to identify the differentially expressed mRNAs after GDF15-EVs treatment. Results GDF15-EVs inhibited apoptosis and promoted autophagy in H2O2 injured H9C2 cells. GDF15-EVs effectively decreased the infarct area and enhanced the cardiac function in rats with AMI. Moreover, GDF15-EVs hindered inflammatory cell infiltration, inhibited cell apoptosis, and promoted cardiac angiogenesis in rats with AMI. RNA sequence showed that telomerase reverse transcriptase (TERT) mRNA was upregulated in GDF15-EVs-treated H9C2 cells. AMPK signaling was activated after GDF15-EVs. Silencing TERT impaired the protective effects of GDF15-EVs on H2O2-injured H9C2 cells. Conclusion GDF15-EVs could fulfil their protective effects against myocardial injury by upregulating the expression of TERT and activating the AMPK signaling pathway. GDF15-EVs might be exploited to design new therapies for AMI.
Collapse
Affiliation(s)
- Ailin Zou
- Department of Cardiology, the Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou, Jiangsu, People’s Republic of China
| | - Tingting Xiao
- Department of Cardiology, the Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou, Jiangsu, People’s Republic of China
| | - Boyu Chi
- Department of Cardiology, the Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou, Jiangsu, People’s Republic of China
- Changzhou Clinical Medical College, Dalian Medical University, Dalian, Liaoning, People’s Republic of China
| | - Yu Wang
- Department of Cardiology, the Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou, Jiangsu, People’s Republic of China
| | - Lipeng Mao
- Department of Cardiology, the Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou, Jiangsu, People’s Republic of China
- Changzhou Clinical Medical College, Dalian Medical University, Dalian, Liaoning, People’s Republic of China
| | - Dabei Cai
- Department of Cardiology, the Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou, Jiangsu, People’s Republic of China
- Changzhou Clinical Medical College, Dalian Medical University, Dalian, Liaoning, People’s Republic of China
| | - Qingqing Gu
- Department of Cardiology, the Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou, Jiangsu, People’s Republic of China
| | - Qianwen Chen
- Department of Cardiology, the Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou, Jiangsu, People’s Republic of China
| | - Qingjie Wang
- Department of Cardiology, the Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou, Jiangsu, People’s Republic of China
| | - Yuan Ji
- Department of Cardiology, the Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou, Jiangsu, People’s Republic of China
| | - Ling Sun
- Department of Cardiology, the Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou, Jiangsu, People’s Republic of China
- Changzhou Clinical Medical College, Dalian Medical University, Dalian, Liaoning, People’s Republic of China
| |
Collapse
|
15
|
Das A, Nikhil A, Shiekh PA, Yadav B, Jagavelu K, Kumar A. Ameliorating impaired cardiac function in myocardial infarction using exosome-loaded gallic-acid-containing polyurethane scaffolds. Bioact Mater 2024; 33:324-340. [PMID: 38076649 PMCID: PMC10701288 DOI: 10.1016/j.bioactmat.2023.11.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 11/06/2023] [Accepted: 11/16/2023] [Indexed: 06/21/2024] Open
Abstract
Myocardial infarction (MI) can be tackled by implanting cardiac patches which provide mechanical support to the heart. However, most tissue-engineered scaffolds face difficulty in attenuating oxidative stress, maintaining mechanical stability, and regenerating damaged cardiomyocytes. Here, we fabricated elastic cryogels using polyurethane modified with antioxidant gallic acid in its backbone (PUGA) and further coated them with decellularized extracellular matrix (dECM) to improve adhesiveness, biocompatibility and hemocompatibility. The scaffold was functionalized with exosomes (EXO) isolated from adipose-derived stem cells having regenerative potential. PUGA-dECM + EXO was tested in a rat model with induced MI where echocardiography after 8 weeks of implantation showed significant recovery in treatment group. Histological analysis revealed a decrease in fibrosis after application of patch and promotion of angiogenesis with reduced oxidative stress was shown by immunostaining. Expression of cardiac tissue contractile function marker was also observed in treatment groups. Thus, the proposed biomaterial has a promising application to be utilized as a patch for cardiac regeneration. More detailed studies with larger animal species are needed for using these observations for specific applications.
Collapse
Affiliation(s)
- Ankita Das
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, 208016, U.P., India
| | - Aman Nikhil
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, 208016, U.P., India
| | - Parvaiz Ahmad Shiekh
- SMART Lab, Centre for Biomedical Engineering, Indian Institute of Technology Delhi, Hauz Khas, New Delhi 110016, India
| | - Babita Yadav
- Department of Pharmacology, Council of Scientific and Industrial Research (CSIR)-Central Drug Research Institute, Lucknow, 226031, U.P., India
| | - Kumaravelu Jagavelu
- Department of Pharmacology, Council of Scientific and Industrial Research (CSIR)-Central Drug Research Institute, Lucknow, 226031, U.P., India
| | - Ashok Kumar
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, 208016, U.P., India
- Centre for Environmental Sciences and Engineering, Indian Institute of Technology Kanpur, Kanpur, 208016, U.P., India
- Centre of Excellence for Orthopaedics and Prosthetics, Gangwal School of Medical Sciences and Technology, Indian Institute of Technology Kanpur, Kanpur, 208016, U.P., India
- The Mehta Family Centre for Engineering in Medicine, Indian Institute of Technology Kanpur, Kanpur, 208016, U.P., India
| |
Collapse
|
16
|
Wan R, Liu S, Feng X, Luo W, Zhang H, Wu Y, Chen S, Shang X. The Revolution of exosomes: From biological functions to therapeutic applications in skeletal muscle diseases. J Orthop Translat 2024; 45:132-139. [PMID: 38544740 PMCID: PMC10966453 DOI: 10.1016/j.jot.2024.01.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 12/29/2023] [Accepted: 01/08/2024] [Indexed: 11/11/2024] Open
Abstract
Skeletal muscle diseases, a broad category encompassing a myriad of afflictions such as acute muscle injury and muscular dystrophies, pose a significant health burden globally. These conditions often lead to muscle weakness, compromised mobility, and a diminished quality of life. In light of this, innovative and effective therapeutic strategies are fervently sought after. Exosomes, naturally extracellular vesicles with a diameter of 30-150 nm, pervade biological fluids. These microscopic entities harbor a host of biological molecules, including proteins, nucleic acids, and lipids, bearing a significant resemblance to their parent cells. The roles they play in the biological theater are manifold, influencing crucial physiological and pathological processes within the organism. In the context of skeletal muscle diseases, their potential extends beyond these roles, as they present a promising therapeutic target and a vehicle for targeted drug delivery. This potentially paves the way for significant clinical applications. This review aims to elucidate the mechanisms underpinning exosome action, their myriad biological functions, and the strides made in exosome research and application. A comprehensive exploration of the part played by exosomes in skeletal muscle repair and regeneration is undertaken. In addition, we delve into the use of exosomes in the therapeutic landscape of skeletal muscle diseases, providing a valuable reference for a deeper understanding of exosome applications in this realm. The concluding section encapsulates the prospective avenues for exosome research and the promising future they hold, underscoring the tremendous potential these diminutive vesicles possess in the field of skeletal muscle diseases. The Translational Potential of this Article. The comprehensive exploration of exosome's diverse biological functions and translational potential in the context of skeletal muscle diseases presented in this review underscores their promising future as a therapeutic target with significant clinical applications, thus paving the way for innovative and effective therapeutic strategies in this realm.
Collapse
Affiliation(s)
- Renwen Wan
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Shan Liu
- Department of Endocrinology, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Xinting Feng
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Wei Luo
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Hanli Zhang
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Yang Wu
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Shiyi Chen
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Xiliang Shang
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, China
| |
Collapse
|
17
|
Xue Y, Riva N, Zhao L, Shieh JS, Chin YT, Gatt A, Guo JJ. Recent advances of exosomes in soft tissue injuries in sports medicine: A critical review on biological and biomaterial applications. J Control Release 2023; 364:90-108. [PMID: 37866405 DOI: 10.1016/j.jconrel.2023.10.031] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 10/08/2023] [Accepted: 10/19/2023] [Indexed: 10/24/2023]
Abstract
Sports medicine is generally associated with soft tissue injuries including muscle injuries, meniscus and ligament injuries, tendon ruptures, tendinopathy, rotator cuff tears, and tendon-bone healing during injuries. Tendon and ligament injuries are the most common sport injuries accounting for 30-40% of all injuries. Therapies for tendon injuries can be divided into surgical and non-surgical methods. Surgical methods mainly depend on the operative procedures, the surgeons and postoperative interventions. In non-surgical methods, cell therapy with stem cells and cell-free therapy with secretome of stem cell origin are current directions. Exosomes are the main paracrine factors of mesenchymal stem cells (MSCs) containing biological components such as proteins, nucleic acids and lipids. Compared with MSCs, MSC-exosomes (MSC-exos) possess the capacity to escape phagocytosis and achieve long-term circulation. In addition, the functions of exosomes from various cell sources in soft tissue injuries in sports medicine have been gradually revealed in recent years. Along with the biological and biomaterial advances in exosomes, exosomes can be designed as drug carriers with biomaterials and exosome research is providing promising contributions in cell biology. Exosomes with biomaterial have the potential of becoming one of the novel therapeutic modalities in regenerative researches. This review summarizes the derives of exosomes in soft tissue regeneration and focuses on the biological and biomaterial mechanism and advances in exosomal therapy in soft tissue injuries.
Collapse
Affiliation(s)
- Yulun Xue
- Department of Orthopaedic Surgery, Suzhou Municipal Hospital/The Affiliated Suzhou Hospital of Nanjing Medical University, Gusu School, Nanjing Medical University, Suzhou 215006, Jiangsu, PR China; Department of Orthopedics and Sports Medicine, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu, PR China
| | - Nicoletta Riva
- Department of Pathology, Faculty of Medicine and Surgery, University of Malta, Msida, Malta
| | - Lingying Zhao
- Jiangsu Institute of Hematology, Key Laboratory of Thrombosis and Hemostasis of Ministry of Health of PR China, Suzhou 215006, Jiangsu, PR China; Department of Hematology, National Clinical Research Center for Hematologic Disease, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu, PR China
| | - Ju-Sheng Shieh
- Department of Periodontology, School of Dentistry, Tri-Service General Hospital, National Defense Medical Center, Taipei City 11490, Taiwan
| | - Yu-Tang Chin
- Department of Periodontology, School of Dentistry, Tri-Service General Hospital, National Defense Medical Center, Taipei City 11490, Taiwan
| | - Alexander Gatt
- Department of Pathology, Faculty of Medicine and Surgery, University of Malta, Msida, Malta; Department of Haematology, Mater Dei Hospital, Msida, Malta
| | - Jiong Jiong Guo
- Department of Orthopedics and Sports Medicine, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu, PR China; Department of Hematology, National Clinical Research Center for Hematologic Disease, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu, PR China.
| |
Collapse
|
18
|
Song Y, You Y, Xu X, Lu J, Huang X, Zhang J, Zhu L, Hu J, Wu X, Xu X, Tan W, Du Y. Adipose-Derived Mesenchymal Stem Cell-Derived Exosomes Biopotentiated Extracellular Matrix Hydrogels Accelerate Diabetic Wound Healing and Skin Regeneration. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2304023. [PMID: 37712174 PMCID: PMC10602544 DOI: 10.1002/advs.202304023] [Citation(s) in RCA: 64] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 07/30/2023] [Indexed: 09/16/2023]
Abstract
Wound healing is an urgent clinical challenge, particularly in the case of chronic wounds. Traditional approaches to wound healing have limited therapeutic efficacy due to lengthy healing times, risk of immune rejection, and susceptibility to infection. Recently, adipose-derived mesenchymal stem cell-derived exosomes (ADSC-exos) have emerged as a promising modality for tissue regeneration and wound repair. In this study, the development of a novel extracellular matrix hydrogel@exosomes (ECM@exo) is reported, which entails incorporation of ADSC-exos into an extracellular matrix hydrogel (ECM hydrogel). This solution forms a hydrogel at physiological temperature (≈37 °C) upon local injection into the wound site. ECM@exo enables sustained release of ADSC-exos from the ECM hydrogel, which maintains high local concentrations at the wound site. The ECM hydrogel displays good biocompatibility and biodegradability. The in vivo and in vitro results demonstrate that ECM@exo treatment effectively reduces inflammation and promotes angiogenesis, collagen deposition, cell proliferation, and migration, thereby accelerating the wound healing process. Overall, this innovative therapeutic approach offers a new avenue for wound healing via a biological hydrogel with controlled exosome release.
Collapse
Affiliation(s)
- Yanling Song
- Institute of PharmaceuticsCollege of Pharmaceutical SciencesZhejiang UniversityHangzhouZhejiang310058P. R. China
| | - Yuchan You
- Institute of PharmaceuticsCollege of Pharmaceutical SciencesZhejiang UniversityHangzhouZhejiang310058P. R. China
| | - Xinyi Xu
- Institute of PharmaceuticsCollege of Pharmaceutical SciencesZhejiang UniversityHangzhouZhejiang310058P. R. China
| | - Jingyi Lu
- Institute of PharmaceuticsCollege of Pharmaceutical SciencesZhejiang UniversityHangzhouZhejiang310058P. R. China
| | - Xiajie Huang
- Institute of PharmaceuticsCollege of Pharmaceutical SciencesZhejiang UniversityHangzhouZhejiang310058P. R. China
| | - Jucong Zhang
- Institute of PharmaceuticsCollege of Pharmaceutical SciencesZhejiang UniversityHangzhouZhejiang310058P. R. China
| | - Luwen Zhu
- Institute of PharmaceuticsCollege of Pharmaceutical SciencesZhejiang UniversityHangzhouZhejiang310058P. R. China
| | - Jiahao Hu
- Institute of PharmaceuticsCollege of Pharmaceutical SciencesZhejiang UniversityHangzhouZhejiang310058P. R. China
| | - Xiaochuan Wu
- Institute of PharmaceuticsCollege of Pharmaceutical SciencesZhejiang UniversityHangzhouZhejiang310058P. R. China
| | - Xiaoling Xu
- Shulan International Medical CollegeZhejiang Shuren UniversityHangzhouZhejiang310015P. R. China
| | - Weiqiang Tan
- Department of Plastic SurgerySir Run Run Shaw HospitalSchool of MedicineZhejiang UniversityHangzhouZhejiang310016P. R. China
| | - Yongzhong Du
- Institute of PharmaceuticsCollege of Pharmaceutical SciencesZhejiang UniversityHangzhouZhejiang310058P. R. China
- Department of Plastic SurgerySir Run Run Shaw HospitalSchool of MedicineZhejiang UniversityHangzhouZhejiang310016P. R. China
- Department of PharmacySir Run Run Shaw HospitalSchool of MedicineZhejiang UniversityHangzhouZhejiang310016P. R. China
- Innovation Center of Translational PharmacyJinhua Institute of Zhejiang UniversityJinhua321299P. R. China
| |
Collapse
|
19
|
Guo J, Yang X, Chen J, Wang C, Sun Y, Yan C, Ren S, Xiong H, Xiang K, Zhang M, Li C, Jiang G, Xiang X, Wan G, Jiang T, Kang Y, Xu X, Chen Z, Li W. Exosomal miR-125b-5p derived from adipose-derived mesenchymal stem cells enhance diabetic hindlimb ischemia repair via targeting alkaline ceramidase 2. J Nanobiotechnology 2023; 21:189. [PMID: 37308908 DOI: 10.1186/s12951-023-01954-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 06/03/2023] [Indexed: 06/14/2023] Open
Abstract
INTRODUCTION Ischemic diseases caused by diabetes continue to pose a major health challenge and effective treatments are in high demand. Mesenchymal stem cells (MSCs) derived exosomes have aroused broad attention as a cell-free treatment for ischemic diseases. However, the efficacy of exosomes from adipose-derived mesenchymal stem cells (ADSC-Exos) in treating diabetic lower limb ischemic injury remains unclear. METHODS Exosomes were isolated from ADSCs culture supernatants by differential ultracentrifugation and their effect on C2C12 cells and HUVECs was assessed by EdU, Transwell, and in vitro tube formation assays separately. The recovery of limb function after ADSC-Exos treatment was evaluated by Laser-Doppler perfusion imaging, limb function score, and histological analysis. Subsequently, miRNA sequencing and rescue experiments were performed to figure out the responsible miRNA for the protective role of ADSC-Exos on diabetic hindlimb ischemic injury. Finally, the direct target of miRNA in C2C12 cells was confirmed by bioinformatic analysis and dual-luciferase report gene assay. RESULTS ADSC-Exos have the potential to promote proliferation and migration of C2C12 cells and to promote HUVECs angiogenesis. In vivo experiments have shown that ADSC-Exos can protect ischemic skeletal muscle, promote the repair of muscle injury, and accelerate vascular regeneration. Combined with bioinformatics analysis, miR-125b-5p may be a key molecule in this process. Transfer of miR-125b-5p into C2C12 cells was able to promote cell proliferation and migration by suppressing ACER2 overexpression. CONCLUSION The findings revealed that miR-125b-5p derived from ADSC-Exos may play a critical role in ischemic muscle reparation by targeting ACER2. In conclusion, our study may provide new insights into the potential of ADSC-Exos as a treatment option for diabetic lower limb ischemia.
Collapse
Affiliation(s)
- Jiahe Guo
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xiaofan Yang
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Jing Chen
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Cheng Wang
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yue Sun
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, 430022, China
| | - Chengqi Yan
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Sen Ren
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Hewei Xiong
- Department of Emergency Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Kaituo Xiang
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Maojie Zhang
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Chengcheng Li
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Guoyong Jiang
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xuejiao Xiang
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Gui Wan
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Tao Jiang
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yu Kang
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xiang Xu
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Zhenbing Chen
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Wenqing Li
- Department of Hand and Foot Surgery, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, 518052, China.
| |
Collapse
|
20
|
Zhang E, Phan P, Zhao Z. Cellular nanovesicles for therapeutic immunomodulation: A perspective on engineering strategies and new advances. Acta Pharm Sin B 2023; 13:1789-1827. [PMID: 37250173 PMCID: PMC10213819 DOI: 10.1016/j.apsb.2022.08.020] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 07/11/2022] [Accepted: 07/28/2022] [Indexed: 02/08/2023] Open
Abstract
Cellular nanovesicles which are referred to as cell-derived, nanosized lipid bilayer structures, have emerged as a promising platform for regulating immune responses. Owing to their outstanding advantages such as high biocompatibility, prominent structural stability, and high loading capacity, cellular nanovesicles are suitable for delivering various immunomodulatory molecules, such as small molecules, nucleic acids, peptides, and proteins. Immunomodulation induced by cellular nanovesicles has been exploited to modulate immune cell behaviors, which is considered as a novel cell-free immunotherapeutic strategy for the prevention and treatment of diverse diseases. Here we review emerging concepts and new advances in leveraging cellular nanovesicles to activate or suppress immune responses, with the aim to explicate their applications for immunomodulation. We overview the general considerations and principles for the design of engineered cellular nanovesicles with tailored immunomodulatory activities. We also discuss new advances in engineering cellular nanovesicles as immunotherapies for treating major diseases.
Collapse
Affiliation(s)
- Endong Zhang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois Chicago, Chicago, IL 60612, USA
| | - Philana Phan
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois Chicago, Chicago, IL 60612, USA
| | - Zongmin Zhao
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois Chicago, Chicago, IL 60612, USA
- Translational Oncology Program, University of Illinois Cancer Center, Chicago, IL 60612, USA
| |
Collapse
|
21
|
Mahindran E, Wan Kamarul Zaman WS, Ahmad Amin Noordin KB, Tan YF, Nordin F. Mesenchymal Stem Cell-Derived Extracellular Vesicles: Hype or Hope for Skeletal Muscle Anti-Frailty. Int J Mol Sci 2023; 24:ijms24097833. [PMID: 37175537 PMCID: PMC10178115 DOI: 10.3390/ijms24097833] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 04/17/2023] [Accepted: 04/18/2023] [Indexed: 05/15/2023] Open
Abstract
Steadily rising population ageing is a global demographic trend due to the advancement of new treatments and technologies in the medical field. This trend also indicates an increasing prevalence of age-associated diseases, such as loss of muscle mass (sarcopenia), which tends to afflict the older population. The deterioration in muscle function can cause severe disability and seriously affects a patient's quality of life. Currently, there is no treatment to prevent and reverse age-related skeletal muscle ageing frailty. Existing interventions mainly slow down and control the signs and symptoms. Mesenchymal stem cell-derived extracellular vesicle (MSC-EV) therapy is a promising approach to attenuate age-related skeletal muscle ageing frailty. However, more studies, especially large-scale randomised clinical trials need to be done in order to determine the adequacy of MSC-EV therapy in treating age-related skeletal muscle ageing frailty. This review compiles the present knowledge of the causes and changes regarding skeletal muscle ageing frailty and the potential of MSC-EV transplantation as a regenerative therapy for age-related skeletal muscle ageing frailty and its clinical trials.
Collapse
Affiliation(s)
- Elancheleyen Mahindran
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, Bandar Tun Razak, Cheras, Kuala Lumpur 56000, Malaysia
| | | | | | - Yuen-Fen Tan
- PPUKM-MAKNA Cancer Center, Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaacob Latif, Bandar Tun Razak, Cheras, Kuala Lumpur 56000, Malaysia
- Faculty of Medicine and Health Sciences, Universiti Tunku Abdul Rahman, Sungai Long Campus, Bandar Sungai Long, Kajang 43000, Malaysia
| | - Fazlina Nordin
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, Bandar Tun Razak, Cheras, Kuala Lumpur 56000, Malaysia
| |
Collapse
|
22
|
Brown PA, Brown PD. Extracellular vesicles and atherosclerotic peripheral arterial disease. Cardiovasc Pathol 2023; 63:107510. [PMID: 36460259 DOI: 10.1016/j.carpath.2022.107510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Revised: 11/24/2022] [Accepted: 11/27/2022] [Indexed: 12/03/2022] Open
Abstract
Atherogenesis involves a complex multifactorial process including chronic inflammation that requires the participation of several cell types and molecules. In addition to their role in vascular homeostasis, extracellular vesicles also appear to play an important role in atherogenesis, including monocyte transmigration and foam cell formation, SMC proliferation and migration, leukocyte transmigration, and thrombosis. Peripheral arterial disease, a major form of peripheral vascular disease, is characterized by structural or functional impairment of peripheral arterial supply, often secondary to atherosclerosis. Elevated levels of extracellular vesicles have been demonstrated in patients with peripheral arterial disease and implicated in the development of atherosclerosis within peripheral vascular beds. However, extracellular vesicles also appear capable of delivering cargo with atheroprotective effects. This capability has been exploited in vesicles engineered to carry content capable of neovascularization, suggesting potential for therapeutic angiogenesis. This dual capacity holds substantial promise for diagnosis and therapy, including possibly limb- and life-saving options for peripheral arterial disease management.
Collapse
Affiliation(s)
- Paul A Brown
- Department of Basic Medical Sciences, University of the West Indies, Mona, Jamaica.
| | - Paul D Brown
- Department of Basic Medical Sciences, University of the West Indies, Mona, Jamaica
| |
Collapse
|
23
|
Zhao C, Xing Z, Wei X, Liao G, Yang D, Liu H, Fan Y. Multibiofunctional TFEB-engineered endothelial progenitor cell-derived extracellular vesicles/hydrogel system for rescuing critical limb ischemia. CHEMICAL ENGINEERING JOURNAL 2023; 460:141730. [DOI: 10.1016/j.cej.2023.141730] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2025]
|
24
|
The effectiveness of cell-derived exosome therapy for diabetic wound: A systematic review and meta-analysis. Ageing Res Rev 2023; 85:101858. [PMID: 36669689 DOI: 10.1016/j.arr.2023.101858] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 01/12/2023] [Accepted: 01/16/2023] [Indexed: 01/19/2023]
Abstract
BACKGROUND The prevalence of diabetes among the elderly population is significant and rising annually. One of the most severe and frequent complications of diabetes mellitus is the diabetic wound, which has long-term negative effects on patients' finances, mental health, and functional abilities. Exosomes, a cell-free therapy, have emerged as a promising novel treatment for diabetic wounds, but their mechanism is still not entirely understood. Therefore, we conducted this meta-analysis to assess the effectiveness of exosomes in the management of diabetic wounds. METHODS We searched PubMed, the Cochrane Library, EMBASE, and Web of Science for pertinent studies that described the therapeutic benefits of exosomes on diabetic wound models that were released before October 17, 2022. The outcome indicators consisted of wound healing rate, neovascular density, re-epithelialization rate, collagen deposition, scar width, and inflammatory factors. RevMan 5.4 software was used to conduct all statistical analyses. RESULTS A total of 21 studies with 323 animals were identified in this meta-analysis. Pooled analyses demonstrated that exosome therapy was shown to be superior to control therapy in terms of wound healing rate (SMD = 5.42; 95 %CI = 4.40-6.44; P < 0.00001), neovascular density (SMD = 5.48; 95 %CI = 4.31-6.64; P < 0.00001), re-epithelialization rate (SMD = 5.06; 95 %CI = 3.75-6.37; P < 0.00001), collagen deposition (SMD = 4.78; 95 %CI = 3.58-5.98; P < 0.00001), scar width (SMD = -8.10; 95 %CI = -10.31 to -5.89; P < 0.00001). Additionally, the expression of inflammatory factors was significantly downregulated in the exosome treatment group. CONCLUSIONS According to this meta-analysis of the current trials, exosome therapy can enhance the quality of diabetic wounds, especially when used in conjunction with novel dressings. To demonstrate the most efficient exosomes and therapeutic parameters for the treatment of diabetic wounds, future studies should conduct sizable, randomized, double-blind trials with high-quality, long-term follow-ups.
Collapse
|
25
|
Huerta CT, Voza FA, Ortiz YY, Liu ZJ, Velazquez OC. Mesenchymal stem cell-based therapy for non-healing wounds due to chronic limb-threatening ischemia: A review of preclinical and clinical studies. Front Cardiovasc Med 2023; 10:1113982. [PMID: 36818343 PMCID: PMC9930203 DOI: 10.3389/fcvm.2023.1113982] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 01/12/2023] [Indexed: 02/04/2023] Open
Abstract
Progressive peripheral arterial disease (PAD) can result in chronic limb-threatening ischemia (CLTI) characterized by clinical complications including rest pain, gangrene and tissue loss. These complications can propagate even more precipitously in the setting of common concomitant diseases in patients with CLTI such as diabetes mellitus (DM). CLTI ulcers are cutaneous, non-healing wounds that persist due to the reduced perfusion and dysfunctional neovascularization associated with severe PAD. Existing therapies for CLTI are primarily limited to anatomic revascularization and medical management of contributing factors such as atherosclerosis and glycemic control. However, many patients fail these treatment strategies and are considered "no-option," thereby requiring extremity amputation, particularly if non-healing wounds become infected or fulminant gangrene develops. Given the high economic burden imposed on patients, decreased quality of life, and poor survival of no-option CLTI patients, regenerative therapies aimed at neovascularization to improve wound healing and limb salvage hold significant promise. Cell-based therapy, specifically utilizing mesenchymal stem/stromal cells (MSCs), is one such regenerative strategy to stimulate therapeutic angiogenesis and tissue regeneration. Although previous reviews have focused primarily on revascularization outcomes after MSC treatments of CLTI with less attention given to their effects on wound healing, here we review advances in pre-clinical and clinical studies related to specific effects of MSC-based therapeutics upon ischemic non-healing wounds associated with CLTI.
Collapse
Affiliation(s)
- Carlos Theodore Huerta
- DeWitt Daughtry Family Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Francesca A. Voza
- DeWitt Daughtry Family Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Yulexi Y. Ortiz
- DeWitt Daughtry Family Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Zhao-Jun Liu
- DeWitt Daughtry Family Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, United States,Vascular Biology Institute, University of Miami Miller School of Medicine, Miami, FL, United States,*Correspondence: Omaida C. Velazquez, ; Zhao-Jun Liu,
| | - Omaida C. Velazquez
- DeWitt Daughtry Family Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, United States,Vascular Biology Institute, University of Miami Miller School of Medicine, Miami, FL, United States,*Correspondence: Omaida C. Velazquez, ; Zhao-Jun Liu,
| |
Collapse
|
26
|
Extracellular Vesicles in Regenerative Processes Associated with Muscle Injury Recovery of Professional Athletes Undergoing Sub Maximal Strength Rehabilitation. Int J Mol Sci 2022; 23:ijms232314913. [PMID: 36499243 PMCID: PMC9739739 DOI: 10.3390/ijms232314913] [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: 10/19/2022] [Revised: 11/21/2022] [Accepted: 11/25/2022] [Indexed: 11/30/2022] Open
Abstract
Platelet-rich plasma (PRP) has great potential in regenerative medicine. In addition to the well-known regenerative potential of secreted growth factors, extracellular vesicles (EVs) are emerging as potential key players in the regulation of tissue repair. However, little is known about their therapeutic potential as regenerative agents. In this study, we have identified and subtyped circulating EVs (platelet-, endothelial-, and leukocyte-derived EVs) in the peripheral blood of athletes recovering from recent muscular injuries and undergoing a submaximal strength rehabilitation program. We found a significant increase in circulating platelet-derived EVs at the end of the rehabilitation program. Moreover, EVs from PRP samples were isolated by fluorescence-activated cell sorting and analyzed by label-free proteomics. The proteomic analysis of PRP-EVs revealed that 32% of the identified proteins were associated to "defense and immunity", and altogether these proteins were involved in vesicle-mediated transport (GO: 0016192; FDR = 3.132 × 10-19), as well as in wound healing (GO: 0042060; FDR = 4.252 × 10-13) and in the events regulating such a process (GO: 0061041; FDR = 2.812 × 10-12). Altogether, these data suggest that platelet-derived EVs may significantly contribute to the regeneration potential of PRP preparations.
Collapse
|
27
|
Sanz-Ros J, Mas-Bargues C, Romero-García N, Huete-Acevedo J, Dromant M, Borrás C. Therapeutic Potential of Extracellular Vesicles in Aging and Age-Related Diseases. Int J Mol Sci 2022; 23:ijms232314632. [PMID: 36498960 PMCID: PMC9735639 DOI: 10.3390/ijms232314632] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 11/21/2022] [Accepted: 11/22/2022] [Indexed: 11/25/2022] Open
Abstract
Aging is associated with an alteration of intercellular communication. These changes in the extracellular environment contribute to the aging phenotype and have been linked to different aging-related diseases. Extracellular vesicles (EVs) are factors that mediate the transmission of signaling molecules between cells. In the aging field, these EVs have been shown to regulate important aging processes, such as oxidative stress or senescence, both in vivo and in vitro. EVs from healthy cells, particularly those coming from stem cells (SCs), have been described as potential effectors of the regenerative potential of SCs. Many studies with different animal models have shown promising results in the field of regenerative medicine. EVs are now viewed as a potential cell-free therapy for tissue damage and several diseases. Here we propose EVs as regulators of the aging process, with an important role in tissue regeneration and a raising therapy for age-related diseases.
Collapse
Affiliation(s)
- Jorge Sanz-Ros
- Freshage Research Group, Department of Physiology, Faculty of Medicine, University of Valencia, Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable-Instituto de Salud Carlos III (CIBERFES-ISCIII), INCLIVA, 46010 Valencia, Spain
- Cardiology Department, Hospital Universitari i Politècnic La Fe, 46026 Valencia, Spain
| | - Cristina Mas-Bargues
- Freshage Research Group, Department of Physiology, Faculty of Medicine, University of Valencia, Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable-Instituto de Salud Carlos III (CIBERFES-ISCIII), INCLIVA, 46010 Valencia, Spain
| | - Nekane Romero-García
- Freshage Research Group, Department of Physiology, Faculty of Medicine, University of Valencia, Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable-Instituto de Salud Carlos III (CIBERFES-ISCIII), INCLIVA, 46010 Valencia, Spain
| | - Javier Huete-Acevedo
- Freshage Research Group, Department of Physiology, Faculty of Medicine, University of Valencia, Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable-Instituto de Salud Carlos III (CIBERFES-ISCIII), INCLIVA, 46010 Valencia, Spain
| | - Mar Dromant
- Freshage Research Group, Department of Physiology, Faculty of Medicine, University of Valencia, Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable-Instituto de Salud Carlos III (CIBERFES-ISCIII), INCLIVA, 46010 Valencia, Spain
| | - Consuelo Borrás
- Freshage Research Group, Department of Physiology, Faculty of Medicine, University of Valencia, Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable-Instituto de Salud Carlos III (CIBERFES-ISCIII), INCLIVA, 46010 Valencia, Spain
- Correspondence:
| |
Collapse
|
28
|
Ji S, Ma P, Cao X, Wang J, Yu X, Luo X, Lu J, Hou W, Zhang Z, Yan Y, Dong Y, Wang H. Myoblast-derived exosomes promote the repair and regeneration of injured skeletal muscle in mice. FEBS Open Bio 2022; 12:2213-2226. [PMID: 36325691 PMCID: PMC9714366 DOI: 10.1002/2211-5463.13504] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 10/17/2022] [Accepted: 11/02/2022] [Indexed: 11/06/2022] Open
Abstract
When skeletal muscle is damaged, satellite cells (SCs) are activated to proliferate rapidly and fuse with the damaged muscle fibers to form new muscle fibers, thereby promoting muscle growth and remodeling and repair of trauma. Exosomes from differentiating human skeletal muscle cells trigger myogenesis of stem cells and provide biochemical cues for skeletal muscle regeneration. Therefore, we hypothesized that, when muscles are injured, myoblast-derived exosomes may regulate muscle repair and regeneration. Here, we investigated the underlying mechanism by applying C2C12-derived exosomes to injured mouse skeletal muscles. The expression levels of skeletal muscle regeneration factors paired box 7 and lipid-promoting factor peroxisome proliferator-activated receptor γ were upregulated, whereas the expression levels of fibrosis factors collagen-1 and α-smooth muscle actin decreased. The expression of proliferating cell nuclear antigen was elevated after applying C2C12-derived exosomes to SCs. Application of C2C12-derived exosomes to fibro-adipogenic progenitors resulted in an increase in peroxisome proliferator-activated receptor γ expression and adipogenesis capacity, whereas α-smooth muscle actin expression and fibrosis capacity decreased. Analysis of the transcriptome and proteome of SCs after treatment with exosomes showed the involvement of multiple biological processes, including proliferation and differentiation of SCs, muscle regeneration, skeletal muscle atrophy, and the inflammatory response after muscle injury. Hence, our data suggest that C2C12-derived exosomes can promote the regeneration of skeletal muscle fibers, accelerate the production of fat from damaged muscles, inhibit the fibrosis of damaged muscles, and accelerate injury repair, which is related to exosome-mediated regulation of the proliferation of SCs, differentiation of fibro-adipogenic progenitors, and modulation of SC mRNA expression and protein formation and decomposition.
Collapse
Affiliation(s)
- Shusen Ji
- College of Veterinary MedicineShanxi Agricultural UniversityJinzhongChina
| | - Pei Ma
- College of Veterinary MedicineShanxi Agricultural UniversityJinzhongChina
| | - Xiaorui Cao
- College of Veterinary MedicineShanxi Agricultural UniversityJinzhongChina
| | - Juan Wang
- Department of Nephrology, Shanghai General HospitalShanghai Jiao Tong University School of MedicineChina
| | - Xiuju Yu
- College of Veterinary MedicineShanxi Agricultural UniversityJinzhongChina
| | - Xiaomao Luo
- College of Veterinary MedicineShanxi Agricultural UniversityJinzhongChina
| | - Jiayin Lu
- College of Veterinary MedicineShanxi Agricultural UniversityJinzhongChina
| | - Wei Hou
- College of Veterinary MedicineShanxi Agricultural UniversityJinzhongChina
| | | | - Yi Yan
- College of Veterinary MedicineShanxi Agricultural UniversityJinzhongChina
| | - Yanjun Dong
- College of Veterinary MedicineChina Agricultural UniversityBeijingChina
| | - Haidong Wang
- College of Veterinary MedicineShanxi Agricultural UniversityJinzhongChina
| |
Collapse
|
29
|
Pan Z, Sun W, Chen Y, Tang H, Lin W, Chen J, Chen C. Extracellular Vesicles in Tissue Engineering: Biology and Engineered Strategy. Adv Healthc Mater 2022; 11:e2201384. [PMID: 36053562 DOI: 10.1002/adhm.202201384] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 08/07/2022] [Indexed: 01/28/2023]
Abstract
Extracellular vesicles (EVs), acting as an important ingredient of intercellular communication through paracrine actions, have gained tremendous attention in the field of tissue engineering (TE). Moreover, these nanosized extracellular particles (30-140 nm) can be incorporated into biomaterials according to different principles to facilitate signal delivery in various regenerative processes directly or indirectly. Bioactive biomaterials as the carrier will extend the retention time and realize the controlled release of EVs, which further enhance their therapeutic efficiency in tissue regeneration. Herein, the basic biological characteristics of EVs are first introduced, and then their outstanding performance in exerting direct impacts on target cells in tissue regeneration as well as indirect effects on promoting angiogenesis and regulating the immune environment, due to specific functional components of EVs (nucleic acid, protein, lipid, etc.), is emphasized. Furthermore, different design ideas for suitable EV-loaded biomaterials are also demonstrated. In the end, this review also highlights the engineered strategies, which aim at solving the problems related to natural EVs such as highly heterogeneous functions, inadequate tissue targeting capabilities, insufficient yield and scalability, etc., thus promoting the therapeutic pertinence and clinical potential of EV-based approaches in TE.
Collapse
Affiliation(s)
- Ziyin Pan
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School Of Medicine, Shanghai, 200092, China.,Shanghai Engineering Research Center of Lung Transplantation, Shanghai, 200433, China
| | - Weiyan Sun
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School Of Medicine, Shanghai, 200092, China.,Shanghai Engineering Research Center of Lung Transplantation, Shanghai, 200433, China
| | - Yi Chen
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School Of Medicine, Shanghai, 200092, China.,Shanghai Engineering Research Center of Lung Transplantation, Shanghai, 200433, China
| | - Hai Tang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School Of Medicine, Shanghai, 200092, China.,Shanghai Engineering Research Center of Lung Transplantation, Shanghai, 200433, China
| | - Weikang Lin
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School Of Medicine, Shanghai, 200092, China.,Shanghai Engineering Research Center of Lung Transplantation, Shanghai, 200433, China
| | - Jiafei Chen
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School Of Medicine, Shanghai, 200092, China
| | - Chang Chen
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School Of Medicine, Shanghai, 200092, China.,Shanghai Engineering Research Center of Lung Transplantation, Shanghai, 200433, China
| |
Collapse
|
30
|
Femminò S, Bonelli F, Brizzi MF. Extracellular vesicles in cardiac repair and regeneration: Beyond stem-cell-based approaches. Front Cell Dev Biol 2022; 10:996887. [PMID: 36120584 PMCID: PMC9479097 DOI: 10.3389/fcell.2022.996887] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 08/16/2022] [Indexed: 11/13/2022] Open
Abstract
The adult human heart poorly regenerate after injury due to the low self-renewal capability retained by adult cardiomyocytes. In the last two decades, several clinical studies have reported the ability of stem cells to induce cardiac regeneration. However, low cell integration and survival into the tissue has limited stem-cell-based clinical approaches. More recently, the release of paracrine mediators including extracellular vesicles (EV) has been recognized as the most relevant mechanism driving benefits upon cell-based therapy. In particular, EV have emerged as key mediators of cardiac repair after damage, in terms of reduction of apoptosis, resolution of inflammation and new blood vessel formation. Herein, mechanisms involved in cardiac damage and regeneration, and current applications of EV and their small non-coding RNAs (miRNAs) in regenerative medicine are discussed.
Collapse
|
31
|
Hao D, Lu L, Song H, Duan Y, Chen J, Carney R, Li JJ, Zhou P, Nolta J, Lam KS, Leach JK, Farmer DL, Panitch A, Wang A. Engineered extracellular vesicles with high collagen-binding affinity present superior in situ retention and therapeutic efficacy in tissue repair. Theranostics 2022; 12:6021-6037. [PMID: 35966577 PMCID: PMC9373818 DOI: 10.7150/thno.70448] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2021] [Accepted: 07/24/2022] [Indexed: 01/26/2023] Open
Abstract
Although stem cell-derived extracellular vesicles (EVs) have remarkable therapeutic potential for various diseases, the therapeutic efficacy of EVs is limited due to their degradation and rapid diffusion after administration, hindering their translational applications. Here, we developed a new generation of collagen-binding EVs, by chemically conjugating a collagen-binding peptide SILY to EVs (SILY-EVs), which were designed to bind to collagen in the extracellular matrix (ECM) and form an EV-ECM complex to improve EVs' in situ retention and therapeutic efficacy after transplantation. Methods: SILY was conjugated to the surface of mesenchymal stem/stromal cell (MSC)-derived EVs by using click chemistry to construct SILY-EVs. Nanoparticle tracking analysis (NTA), ExoView analysis, cryogenic electron microscopy (cryo-EM) and western-blot analysis were used to characterize the SILY-EVs. Fluorescence imaging (FLI), MTS assay, ELISA and reverse transcription-quantitative polymerase chain reaction (RT-qPCR) were used to evaluate the collagen binding and biological functions of SILY-EVs in vitro. In a mouse hind limb ischemia model, the in vivo imaging system (IVIS), laser doppler perfusion imaging (LDPI), micro-CT, FLI and RT-qPCR were used to determine the SILY-EV retention, inflammatory response, blood perfusion, gene expression, and tissue regeneration. Results:In vitro, the SILY conjugation significantly enhanced EV adhesion to the collagen surface and did not alter the EVs' biological functions. In the mouse hind limb ischemia model, SILY-EVs presented longer in situ retention, suppressed inflammatory responses, and significantly augmented muscle regeneration and vascularization, compared to the unmodified EVs. Conclusion: With the broad distribution of collagen in various tissues and organs, SILY-EVs hold promise to improve the therapeutic efficacy of EV-mediated treatment in a wide range of diseases and disorders. Moreover, SILY-EVs possess the potential to functionalize collagen-based biomaterials and deliver therapeutic agents for regenerative medicine applications.
Collapse
Affiliation(s)
- Dake Hao
- Department of Surgery, University of California Davis, Sacramento, CA 95817, USA
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children Northern California, Sacramento, CA 95817, USA
| | - Lu Lu
- Department of Surgery, University of California Davis, Sacramento, CA 95817, USA
| | - Hengyue Song
- Department of Surgery, University of California Davis, Sacramento, CA 95817, USA
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children Northern California, Sacramento, CA 95817, USA
| | - Yixin Duan
- Department of Radiation Oncology, University of California Davis, Sacramento, CA 95817, USA
| | - Jianing Chen
- Department of Surgery, University of California Davis, Sacramento, CA 95817, USA
| | - Randy Carney
- Department of Biomedical Engineering, University of California Davis, Davis, CA 95616, USA
| | - Jian Jian Li
- Department of Radiation Oncology, University of California Davis, Sacramento, CA 95817, USA
| | - Ping Zhou
- Stem Cell Program, Department of Internal Medicine, University of California Davis Medical Center, Sacramento, CA 95817, USA
| | - Jan Nolta
- Stem Cell Program, Department of Internal Medicine, University of California Davis Medical Center, Sacramento, CA 95817, USA
| | - Kit S. Lam
- Department of Biochemistry and Molecular Medicine, University of California Davis, Sacramento, CA 95817, USA
| | - J. Kent Leach
- Department of Orthopaedic Surgery, School of Medicine, University of California Davis, Sacramento, CA 95817, USA
| | - Diana L Farmer
- Department of Surgery, University of California Davis, Sacramento, CA 95817, USA
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children Northern California, Sacramento, CA 95817, USA
| | - Alyssa Panitch
- Department of Surgery, University of California Davis, Sacramento, CA 95817, USA
- Department of Biomedical Engineering, University of California Davis, Davis, CA 95616, USA
| | - Aijun Wang
- Department of Surgery, University of California Davis, Sacramento, CA 95817, USA
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children Northern California, Sacramento, CA 95817, USA
- Department of Biomedical Engineering, University of California Davis, Davis, CA 95616, USA
| |
Collapse
|
32
|
Rome S. Muscle and Adipose Tissue Communicate with Extracellular Vesicles. Int J Mol Sci 2022; 23:ijms23137052. [PMID: 35806052 PMCID: PMC9266961 DOI: 10.3390/ijms23137052] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 06/17/2022] [Accepted: 06/23/2022] [Indexed: 02/05/2023] Open
Abstract
In numerous body locations, muscle and adipose tissue are in close contact. Both tissues are endocrine organs that release cytokines, playing a crutial role in the control of tissue homeostasis in health and diseases. Within this context, the identification of the signals involved in muscle–fat crosstalk has been a hot topic over the last 15 years. Recently, it has been discovered that adipose tissue and muscles can release information embedded in lipid-derived nanovesicles called ‘extracellular vesicles’ (EVs), which can modulate the phenotype and the homeostasis of neighboring recipient cells. This article reviews knowledge on EVs and their involvement in the communication between adipose tissue and muscle in several body locations. Even if the works are scarce, they have revolutionized our vision in the field of metabolic and cardiovascular diseases.
Collapse
Affiliation(s)
- Sophie Rome
- CarMeN Laboratory, INSERM 1060/INRAE 1397, Lyon-Sud Faculty of Medicine, LYON 1 University, 69301 Pierre Bénite, France
| |
Collapse
|
33
|
Li M, Fang F, Sun M, Zhang Y, Hu M, Zhang J. Extracellular vesicles as bioactive nanotherapeutics: An emerging paradigm for regenerative medicine. Theranostics 2022; 12:4879-4903. [PMID: 35836815 PMCID: PMC9274746 DOI: 10.7150/thno.72812] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Accepted: 06/05/2022] [Indexed: 01/12/2023] Open
Abstract
In recent decades, extracellular vesicles (EVs), as bioactive cell-secreted nanoparticles which are involved in various physiological and pathological processes including cell proliferation, immune regulation, angiogenesis and tissue repair, have emerged as one of the most attractive nanotherapeutics for regenerative medicine. Herein we provide a systematic review of the latest progress of EVs for regenerative applications. Firstly, we will briefly introduce the biogenesis, function and isolation technology of EVs. Then, the underlying therapeutic mechanisms of the native unmodified EVs and engineering strategies of the modified EVs as regenerative entities will be discussed. Subsequently, the main focus will be placed on the tissue repair and regeneration applications of EVs on various organs including brain, heart, bone and cartilage, liver and kidney, as well as skin. More importantly, current clinical trials of EVs for regenerative medicine will also be briefly highlighted. Finally, the future challenges and insightful perspectives of the currently developed EV-based nanotherapeutics in biomedicine will be discussed. In short, the bioactive EV-based nanotherapeutics have opened new horizons for biologists, chemists, nanoscientists, pharmacists, as well as clinicians, making possible powerful tools and therapies for regenerative medicine.
Collapse
Affiliation(s)
- Min Li
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Sciences, Beijing Institute of Technology, Beijing 100081, P. R. China
| | - Fang Fang
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Sciences, Beijing Institute of Technology, Beijing 100081, P. R. China
| | - Meng Sun
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Sciences, Beijing Institute of Technology, Beijing 100081, P. R. China
| | - Yinfeng Zhang
- International Medical Center, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, P. R. China
| | - Min Hu
- Department of Hepatobiliary Surgery, Jinan University First Affiliated Hospital, Guangzhou, 510630, P. R. China
| | - Jinfeng Zhang
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Sciences, Beijing Institute of Technology, Beijing 100081, P. R. China
| |
Collapse
|
34
|
Prophylactic Evidence of MSCs-Derived Exosomes in Doxorubicin/Trastuzumab-Induced Cardiotoxicity: Beyond Mechanistic Target of NRG-1/Erb Signaling Pathway. Int J Mol Sci 2022; 23:ijms23115967. [PMID: 35682646 PMCID: PMC9181089 DOI: 10.3390/ijms23115967] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Revised: 05/19/2022] [Accepted: 05/23/2022] [Indexed: 12/06/2022] Open
Abstract
Trastuzumab (Trz) is a humanized monoclonal antibody targeting epidermal growth factor receptor 2 (HER2; ErbB2). The combined administration of Trz and doxorubicin (DOX) has shown potent anti-cancer efficacy; however, this regimen may be accompanied by severe cardiac toxicity. Mesenchymal stem cells (MSCs)-derived exosomes are nanosized vesicles that play a crucial role in cell–cell communication and have shown efficacy in the treatment of various diseases. In this study, we aim to investigate the cardioprotective effects of MSCs-derived exosomes in a DOX/Trz- mediated cardiotoxicity model, and the possible mechanisms underlying these effects are elucidated. Forty-nine male rats were randomly assigned into four groups: Group I (control); Group II (Dox/Trz); Group III (protective group); and Group IV (curative group). Cardiac hemodynamic parameters, serum markers of cardiac injury, oxidative stress indices, and cardiac histopathology were investigated. Further, transcript profile of specific cardiac tissue injury markers, apoptotic markers, and fibrotic markers were analyzed using qRT-PCR, while the protein expressions of pAkt/Akt, pERK/ERK, pJNK/JNK, pJNK/JNK, and pSTAT3/STAT3 were evaluated by ELISA. Additionally, cardiac mirR-21 and miR-26a were assessed. A combined administration of DOX/Trz disrupted redox and Ca2+ homeostasis in cardiac tissue induced myocardial fibrosis and myofibril loss and triggered cardiac DNA damage and apoptosis. This cardiotoxicity was accompanied by decreased NRG-1 mRNA expression, HER2 protein expression, and suppressed AKT and ERK phosphorylation, while triggering JNK phosphorylation. Histological and ultra-structural examination of cardiac specimens revealed features typical of cardiac tissue injury. Moreover, a significant decline in cardiac function was observed through biochemical testing of serum cardiac markers and echocardiography. In contrast, the intraperitoneal administration of MSCs-derived exosomes alleviated cardiac injury in both protective and curative protocols; however, superior effects were observed in the protective protocol. The results of the current study indicate the ability of MSCs-derived exosomes to protect from and attenuate DOX/Trz-induced cardiotoxicity. The NRG-1/HER2, MAPK, PI3K/AKT, PJNK/JNK, and PSTAT/STAT signaling pathways play roles in mediating these effects.
Collapse
|
35
|
Wan R, Hussain A, Behfar A, Moran SL, Zhao C. The Therapeutic Potential of Exosomes in Soft Tissue Repair and Regeneration. Int J Mol Sci 2022; 23:ijms23073869. [PMID: 35409228 PMCID: PMC8998690 DOI: 10.3390/ijms23073869] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 03/28/2022] [Accepted: 03/28/2022] [Indexed: 12/12/2022] Open
Abstract
Soft tissue defects are common following trauma and tumor extirpation. These injuries can result in poor functional recovery and lead to a diminished quality of life. The healing of skin and muscle is a complex process that, at present, leads to incomplete recovery and scarring. Regenerative medicine may offer the opportunity to improve the healing process and functional outcomes. Barriers to regenerative strategies have included cost, regulatory hurdles, and the need for cell-based therapies. In recent years, exosomes, or extracellular vesicles, have gained tremendous attention in the field of soft tissue repair and regeneration. These nanosized extracellular particles (30-140 nm) can break the cellular boundaries, as well as facilitate intracellular signal delivery in various regenerative physiologic and pathologic processes. Existing studies have established the potential of exosomes in regenerating tendons, skeletal muscles, and peripheral nerves through different mechanisms, including promoting myogenesis, increasing tenocyte differentiation and enhancing neurite outgrowth, and the proliferation of Schwann cells. These exosomes can be stored for immediate use in the operating room, and can be produced cost efficiently. In this article, we critically review the current advances of exosomes in soft tissue (tendons, skeletal muscles, and peripheral nerves) healing. Additionally, new directions for clinical applications in the future will be discussed.
Collapse
Affiliation(s)
- Rou Wan
- Division of Plastic Surgery, Mayo Clinic, Rochester, MN 55905, USA; (R.W.); (A.H.); (S.L.M.)
| | - Arif Hussain
- Division of Plastic Surgery, Mayo Clinic, Rochester, MN 55905, USA; (R.W.); (A.H.); (S.L.M.)
| | - Atta Behfar
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN 55905, USA;
- Van Cleve Cardiac Regenerative Medicine Program, Center for Regenerative Medicine, Mayo Clinic, Rochester, MN 55905, USA
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905, USA
| | - Steven L. Moran
- Division of Plastic Surgery, Mayo Clinic, Rochester, MN 55905, USA; (R.W.); (A.H.); (S.L.M.)
| | - Chunfeng Zhao
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN 55905, USA
- Correspondence:
| |
Collapse
|
36
|
The sustained PGE 2 release matrix improves neovascularization and skeletal muscle regeneration in a hindlimb ischemia model. J Nanobiotechnology 2022; 20:95. [PMID: 35209908 PMCID: PMC8867652 DOI: 10.1186/s12951-022-01301-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Accepted: 02/06/2022] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND The promising therapeutic strategy for the treatment of peripheral artery disease (PAD) is to restore blood supply and promote regeneration of skeletal muscle regeneration. Increasing evidence revealed that prostaglandin E2 (PGE2), a lipid signaling molecule, has significant therapeutic potential for tissue repair and regeneration. Though PGE2 has been well reported in tissue regeneration, the application of PGE2 is hampered by its short half-life in vivo and the lack of a viable system for sustained release of PGE2. RESULTS In this study, we designed and synthesized a new PGE2 release matrix by chemically bonding PGE2 to collagen. Our results revealed that the PGE2 matrix effectively extends the half-life of PGE2 in vitro and in vivo. Moreover, the PGE2 matrix markedly improved neovascularization by increasing angiogenesis, as confirmed by bioluminescence imaging (BLI). Furthermore, the PGE2 matrix exhibits superior therapeutic efficacy in the hindlimb ischemia model through the activation of MyoD1-mediated muscle stem cells, which is consistent with accelerated structural recovery of skeletal muscle, as evidenced by histological analysis. CONCLUSIONS Our findings highlight the chemical bonding strategy of chemical bonding PGE2 to collagen for sustained release and may facilitate the development of PGE2-based therapies to significantly improve tissue regeneration.
Collapse
|
37
|
Martin-Ventura JL, Roncal C, Orbe J, Blanco-Colio LM. Role of Extracellular Vesicles as Potential Diagnostic and/or Therapeutic Biomarkers in Chronic Cardiovascular Diseases. Front Cell Dev Biol 2022; 10:813885. [PMID: 35155428 PMCID: PMC8827403 DOI: 10.3389/fcell.2022.813885] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 01/10/2022] [Indexed: 12/12/2022] Open
Abstract
Cardiovascular diseases (CVDs) are the first cause of death worldwide. In recent years, there has been great interest in the analysis of extracellular vesicles (EVs), including exosomes and microparticles, as potential mediators of biological communication between circulating cells/plasma and cells of the vasculature. Besides their activity as biological effectors, EVs have been also investigated as circulating/systemic biomarkers in different acute and chronic CVDs. In this review, the role of EVs as potential diagnostic and prognostic biomarkers in chronic cardiovascular diseases, including atherosclerosis (mainly, peripheral arterial disease, PAD), aortic stenosis (AS) and aortic aneurysms (AAs), will be described. Mechanistically, we will analyze the implication of EVs in pathological processes associated to cardiovascular remodeling, with special emphasis in their role in vascular and valvular calcification. Specifically, we will focus on the participation of EVs in calcium accumulation in the pathological vascular wall and aortic valves, involving the phenotypic change of vascular smooth muscle cells (SMCs) or valvular interstitial cells (IC) to osteoblast-like cells. The knowledge of the implication of EVs in the pathogenic mechanisms of cardiovascular remodeling is still to be completely deciphered but there are promising results supporting their potential translational application to the diagnosis and therapy of different CVDs.
Collapse
Affiliation(s)
- Jose Luis Martin-Ventura
- Vascular Research Laboratory, IIS-Fundación Jiménez-Díaz, Madrid, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
- *Correspondence: Jose Luis Martin-Ventura, ; Carmen Roncal,
| | - Carmen Roncal
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
- Laboratory of Atherothrombosis, Program of Cardiovascular Diseases, Cima Universidad de Navarra, Instituto de Investigación Sanitaria de Navarra, IdiSNA, Pamplona, Spain
- *Correspondence: Jose Luis Martin-Ventura, ; Carmen Roncal,
| | - Josune Orbe
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
- Laboratory of Atherothrombosis, Program of Cardiovascular Diseases, Cima Universidad de Navarra, Instituto de Investigación Sanitaria de Navarra, IdiSNA, Pamplona, Spain
| | - Luis Miguel Blanco-Colio
- Vascular Research Laboratory, IIS-Fundación Jiménez-Díaz, Madrid, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| |
Collapse
|
38
|
Giraud R, Moyon A, Simoncini S, Duchez AC, Nail V, Chareyre C, Bouhlel A, Balasse L, Fernandez S, Vallier L, Hache G, Sabatier F, Dignat-George F, Lacroix R, Guillet B, Garrigue P. Tracking Radiolabeled Endothelial Microvesicles Predicts Their Therapeutic Efficacy: A Proof-of-Concept Study in Peripheral Ischemia Mouse Model Using SPECT/CT Imaging. Pharmaceutics 2022; 14:pharmaceutics14010121. [PMID: 35057018 PMCID: PMC8778059 DOI: 10.3390/pharmaceutics14010121] [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: 12/01/2021] [Revised: 12/24/2021] [Accepted: 12/29/2021] [Indexed: 11/16/2022] Open
Abstract
Microvesicles, so-called endothelial large extracellular vesicles (LEVs), are of great interest as biological markers and cell-free biotherapies in cardiovascular and oncologic diseases. However, their therapeutic perspectives remain limited due to the lack of reliable data regarding their systemic biodistribution after intravenous administration. Methods: Applied to a mouse model of peripheral ischemia, radiolabeled endothelial LEVs were tracked and their in vivo whole-body distribution was quantified by microSPECT/CT imaging. Hindlimb perfusion was followed by LASER Doppler and motility impairment function was evaluated up to day 28 post-ischemia. Results: Early and specific homing of LEVs to ischemic hind limbs was quantified on the day of ischemia and positively correlated with reperfusion intensity at a later stage on day 28 after ischemia, associated with an improved motility function. Conclusions: This concept is a major asset for investigating the biodistribution of LEVs issued from other cell types, including cancer, thus partly contributing to better knowledge and understanding of their fate after injection.
Collapse
Affiliation(s)
- Romain Giraud
- C2VN, INSERM, INRAE, Aix Marseille University, 13385 Marseille, France; (R.G.); (A.M.); (S.S.); (A.-C.D.); (C.C.); (A.B.); (L.B.); (L.V.); (G.H.); (F.S.); (F.D.-G.); (R.L.); (B.G.)
- CERIMED, CNRS, Marseille, Aix Marseille University, 13385 Marseille, France; (V.N.); (S.F.)
- Radiopharmacy, Pôle Pharmacie, University Hospitals of Marseille, APHM, 13005 Marseille, France
| | - Anaïs Moyon
- C2VN, INSERM, INRAE, Aix Marseille University, 13385 Marseille, France; (R.G.); (A.M.); (S.S.); (A.-C.D.); (C.C.); (A.B.); (L.B.); (L.V.); (G.H.); (F.S.); (F.D.-G.); (R.L.); (B.G.)
- CERIMED, CNRS, Marseille, Aix Marseille University, 13385 Marseille, France; (V.N.); (S.F.)
- Radiopharmacy, Pôle Pharmacie, University Hospitals of Marseille, APHM, 13005 Marseille, France
| | - Stéphanie Simoncini
- C2VN, INSERM, INRAE, Aix Marseille University, 13385 Marseille, France; (R.G.); (A.M.); (S.S.); (A.-C.D.); (C.C.); (A.B.); (L.B.); (L.V.); (G.H.); (F.S.); (F.D.-G.); (R.L.); (B.G.)
| | - Anne-Claire Duchez
- C2VN, INSERM, INRAE, Aix Marseille University, 13385 Marseille, France; (R.G.); (A.M.); (S.S.); (A.-C.D.); (C.C.); (A.B.); (L.B.); (L.V.); (G.H.); (F.S.); (F.D.-G.); (R.L.); (B.G.)
| | - Vincent Nail
- CERIMED, CNRS, Marseille, Aix Marseille University, 13385 Marseille, France; (V.N.); (S.F.)
- Radiopharmacy, Pôle Pharmacie, University Hospitals of Marseille, APHM, 13005 Marseille, France
| | - Corinne Chareyre
- C2VN, INSERM, INRAE, Aix Marseille University, 13385 Marseille, France; (R.G.); (A.M.); (S.S.); (A.-C.D.); (C.C.); (A.B.); (L.B.); (L.V.); (G.H.); (F.S.); (F.D.-G.); (R.L.); (B.G.)
| | - Ahlem Bouhlel
- C2VN, INSERM, INRAE, Aix Marseille University, 13385 Marseille, France; (R.G.); (A.M.); (S.S.); (A.-C.D.); (C.C.); (A.B.); (L.B.); (L.V.); (G.H.); (F.S.); (F.D.-G.); (R.L.); (B.G.)
- CERIMED, CNRS, Marseille, Aix Marseille University, 13385 Marseille, France; (V.N.); (S.F.)
| | - Laure Balasse
- C2VN, INSERM, INRAE, Aix Marseille University, 13385 Marseille, France; (R.G.); (A.M.); (S.S.); (A.-C.D.); (C.C.); (A.B.); (L.B.); (L.V.); (G.H.); (F.S.); (F.D.-G.); (R.L.); (B.G.)
- CERIMED, CNRS, Marseille, Aix Marseille University, 13385 Marseille, France; (V.N.); (S.F.)
| | - Samantha Fernandez
- CERIMED, CNRS, Marseille, Aix Marseille University, 13385 Marseille, France; (V.N.); (S.F.)
| | - Loris Vallier
- C2VN, INSERM, INRAE, Aix Marseille University, 13385 Marseille, France; (R.G.); (A.M.); (S.S.); (A.-C.D.); (C.C.); (A.B.); (L.B.); (L.V.); (G.H.); (F.S.); (F.D.-G.); (R.L.); (B.G.)
| | - Guillaume Hache
- C2VN, INSERM, INRAE, Aix Marseille University, 13385 Marseille, France; (R.G.); (A.M.); (S.S.); (A.-C.D.); (C.C.); (A.B.); (L.B.); (L.V.); (G.H.); (F.S.); (F.D.-G.); (R.L.); (B.G.)
- CERIMED, CNRS, Marseille, Aix Marseille University, 13385 Marseille, France; (V.N.); (S.F.)
| | - Florence Sabatier
- C2VN, INSERM, INRAE, Aix Marseille University, 13385 Marseille, France; (R.G.); (A.M.); (S.S.); (A.-C.D.); (C.C.); (A.B.); (L.B.); (L.V.); (G.H.); (F.S.); (F.D.-G.); (R.L.); (B.G.)
| | - Françoise Dignat-George
- C2VN, INSERM, INRAE, Aix Marseille University, 13385 Marseille, France; (R.G.); (A.M.); (S.S.); (A.-C.D.); (C.C.); (A.B.); (L.B.); (L.V.); (G.H.); (F.S.); (F.D.-G.); (R.L.); (B.G.)
- Department of Hematology and Vascular Biology, University Hospitals of Marseille, APHM, 13005 Marseille, France
| | - Romaric Lacroix
- C2VN, INSERM, INRAE, Aix Marseille University, 13385 Marseille, France; (R.G.); (A.M.); (S.S.); (A.-C.D.); (C.C.); (A.B.); (L.B.); (L.V.); (G.H.); (F.S.); (F.D.-G.); (R.L.); (B.G.)
- Department of Hematology and Vascular Biology, University Hospitals of Marseille, APHM, 13005 Marseille, France
| | - Benjamin Guillet
- C2VN, INSERM, INRAE, Aix Marseille University, 13385 Marseille, France; (R.G.); (A.M.); (S.S.); (A.-C.D.); (C.C.); (A.B.); (L.B.); (L.V.); (G.H.); (F.S.); (F.D.-G.); (R.L.); (B.G.)
- CERIMED, CNRS, Marseille, Aix Marseille University, 13385 Marseille, France; (V.N.); (S.F.)
- Radiopharmacy, Pôle Pharmacie, University Hospitals of Marseille, APHM, 13005 Marseille, France
| | - Philippe Garrigue
- C2VN, INSERM, INRAE, Aix Marseille University, 13385 Marseille, France; (R.G.); (A.M.); (S.S.); (A.-C.D.); (C.C.); (A.B.); (L.B.); (L.V.); (G.H.); (F.S.); (F.D.-G.); (R.L.); (B.G.)
- CERIMED, CNRS, Marseille, Aix Marseille University, 13385 Marseille, France; (V.N.); (S.F.)
- Radiopharmacy, Pôle Pharmacie, University Hospitals of Marseille, APHM, 13005 Marseille, France
- Correspondence:
| |
Collapse
|
39
|
Deng H, Chen Y. The role of adipose-derived stem cells-derived extracellular vesicles in the treatment of diabetic foot ulcer: Trends and prospects. Front Endocrinol (Lausanne) 2022; 13:902130. [PMID: 35966086 PMCID: PMC9363610 DOI: 10.3389/fendo.2022.902130] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 07/01/2022] [Indexed: 11/13/2022] Open
Abstract
Diabetic foot ulcer(DFU) is one of the most severe chronic complications of type 2 diabetes mellitus, which is mainly caused by peripheral vascular occlusion with various degrees of infection. Treatment of DFU is difficult, and ulcer formation in lower limbs and deep-tissue necrosis might lead to disability or even death. Insulin resistance is the major mechanism of type 2 diabetes mellitus development, largely caused by adipose tissue dysfunction. However, adipose tissue was recently identified as an important endocrine organ that secretes bio-active factors, such as adipokines and extracellular vesicles(EVs). And adipose tissue-derived stem cells(ADSCs) are abundant in adipose tissue and have become a hot topic in the tissue engineering field. In particular, EVs derived from ADSCs contain abundant biomarkers and mediators. These EVs exert significant effects on distant cells and organs, contributing to metabolic homeostasis. In this review, we aim to elaborate on the mechanisms of diabetic non-healing wound development and the role of ADSCs-EVs in wound repair, which might provide a new therapy for treating DFU.
Collapse
Affiliation(s)
- Hongyan Deng
- Division of Endocrinology, Internal Medicine, Tongji Hospital, Huazhong University of Science & Technology, Wuhan, China
- Laboratory of Endocrinology, Tongji Hospital, Huazhong University of Science & Technology, Wuhan, China
| | - Yong Chen
- Division of Endocrinology, Internal Medicine, Tongji Hospital, Huazhong University of Science & Technology, Wuhan, China
- Laboratory of Endocrinology, Tongji Hospital, Huazhong University of Science & Technology, Wuhan, China
- Branch of National Clinical Research Center for Metabolic Diseases, Hubei, China
- *Correspondence: Yong Chen,
| |
Collapse
|
40
|
Zhang Y, Wang H, Oliveira RHM, Zhao C, Popel AS. Systems biology of angiogenesis signaling: Computational models and omics. WIREs Mech Dis 2021; 14:e1550. [PMID: 34970866 PMCID: PMC9243197 DOI: 10.1002/wsbm.1550] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 12/03/2021] [Accepted: 12/06/2021] [Indexed: 01/10/2023]
Abstract
Angiogenesis is a highly regulated multiscale process that involves a plethora of cells, their cellular signal transduction, activation, proliferation, differentiation, as well as their intercellular communication. The coordinated execution and integration of such complex signaling programs is critical for physiological angiogenesis to take place in normal growth, development, exercise, and wound healing, while its dysregulation is critically linked to many major human diseases such as cancer, cardiovascular diseases, and ocular disorders; it is also crucial in regenerative medicine. Although huge efforts have been devoted to drug development for these diseases by investigation of angiogenesis‐targeted therapies, only a few therapeutics and targets have proved effective in humans due to the innate multiscale complexity and nonlinearity in the process of angiogenic signaling. As a promising approach that can help better address this challenge, systems biology modeling allows the integration of knowledge across studies and scales and provides a powerful means to mechanistically elucidate and connect the individual molecular and cellular signaling components that function in concert to regulate angiogenesis. In this review, we summarize and discuss how systems biology modeling studies, at the pathway‐, cell‐, tissue‐, and whole body‐levels, have advanced our understanding of signaling in angiogenesis and thereby delivered new translational insights for human diseases. This article is categorized under:Cardiovascular Diseases > Computational Models Cancer > Computational Models
Collapse
Affiliation(s)
- Yu Zhang
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Hanwen Wang
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Rebeca Hannah M Oliveira
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Chen Zhao
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Aleksander S Popel
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
41
|
Hong P, Yu M, Tian W. Diverse RNAs in adipose-derived extracellular vesicles and their therapeutic potential. MOLECULAR THERAPY. NUCLEIC ACIDS 2021; 26:665-677. [PMID: 34703651 PMCID: PMC8516999 DOI: 10.1016/j.omtn.2021.08.028] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Adipose tissue, which is considered an energy storage and active endocrine organ, produces and secretes a large amount of adipokines to regulate distant targets through blood circulation, especially extracellular vesicles (EVs). As cell-derived, membranous nanoparticles, EVs have recently garnered great attention as novel mediators in establishing intercellular communications as well as in accelerating interorgan crosstalk. Studies have revealed that the RNAs, including coding RNAs (messenger RNAs) and noncoding RNAs (long noncoding RNAs, microRNAs, and circular RNAs) are key bioactive cargoes of EV functions in various pathophysiological processes, such as cell differentiation, metabolic homeostasis, immune signal transduction, and cancer. Moreover, certain EV-contained RNAs have gradually been recognized as novel biomarkers, prognostic indicators, or even therapeutic nanodrugs of diseases. Therefore, in this review, we comprehensively summarize different classes of RNAs presented in adipose-derived EVs and discuss their therapeutic potential according to the latest research progress to provide valuable knowledge in this area.
Collapse
Affiliation(s)
- Pengyu Hong
- State Key Laboratory of Oral Disease & National Clinical Research Center for Oral Diseases West China School of Stomatology, Sichuan University, Chengdu, China.,National Engineering Laboratory for Oral Regenerative Medicine, West China School of Stomatology, Sichuan University, Chengdu, China.,Engineering Research Center of Oral Translational Medicine, Ministry of Education, Sichuan University, Chengdu, China.,Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Mei Yu
- State Key Laboratory of Oral Disease & National Clinical Research Center for Oral Diseases West China School of Stomatology, Sichuan University, Chengdu, China.,National Engineering Laboratory for Oral Regenerative Medicine, West China School of Stomatology, Sichuan University, Chengdu, China.,Engineering Research Center of Oral Translational Medicine, Ministry of Education, Sichuan University, Chengdu, China
| | - Weidong Tian
- State Key Laboratory of Oral Disease & National Clinical Research Center for Oral Diseases West China School of Stomatology, Sichuan University, Chengdu, China.,National Engineering Laboratory for Oral Regenerative Medicine, West China School of Stomatology, Sichuan University, Chengdu, China.,Engineering Research Center of Oral Translational Medicine, Ministry of Education, Sichuan University, Chengdu, China.,Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
42
|
Exosomes from adipose-derived stem cells alleviate myocardial infarction via microRNA-31/FIH1/HIF-1α pathway. J Mol Cell Cardiol 2021; 162:10-19. [PMID: 34474073 DOI: 10.1016/j.yjmcc.2021.08.010] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 08/13/2021] [Accepted: 08/25/2021] [Indexed: 02/08/2023]
Abstract
Our previous study has revealed that exosomes from adipose-derived stem cells (ASCs) promote angiogenesis in subcutaneously transplanted gels by delivery of microRNA-31 (miR-31) which targets factor inhibiting hypoxia-inducible factor-1 (FIH1) in recipient cells. Here we hypothesized that ASC exosomes alleviate ischemic diseases through miR-31/FIH1/hypoxia-inducible factor-1α (HIF-1α) signaling pathway. Exosomes from ASCs were characterized with nanoparticle tracking analysis, transmission electron microscopy, and immunoblotting analysis for exosomal markers. Results from immunoblotting and laser imaging of ischemic mouse hindlimb revealed that miR-31 enriched ASC exosomes inhibited FIH1 expression and enhanced the blood perfusion, respectively. These effects were impaired when using miR-31-depleted exosomes. Immunohistochemistry analysis showed that administration of exosomes resulted in a higher arteriole density and larger CD31+ area in ischemic hindlimb than miR-31-delpleted exosomes. Similarly, knockdown of miR-31 in exosomes reduced the effects of the exosomes on increasing ventricular fraction shortening and CD31+ area, and on decreasing infarct size. Exosomes promoted endothelial cell migration and tube formation. These changes were attenuated when miR-31 was depleted in the exosomes or when FIH1 was overexpressed in the endothelial cells. Furthermore, the results from immunocytochemistry, co-immunoprecipitation, and luciferase reporter assay demonstrated that the effects of exosomes on nuclear translocation, binding with co-activator p300, and activation of HIF-1α were decreased when miR-31 was depleted in the exosomes or FIH1 was overexpressed. Our findings provide evidence that exosomes from ASCs promote angiogenesis in both mouse ischemic hindlimb and heart through transport of miR-31 which targets FIH1 and therefore triggers HIF-1α transcriptional activation.
Collapse
|
43
|
Nagelkerke A, Ojansivu M, van der Koog L, Whittaker TE, Cunnane EM, Silva AM, Dekker N, Stevens MM. Extracellular vesicles for tissue repair and regeneration: Evidence, challenges and opportunities. Adv Drug Deliv Rev 2021; 175:113775. [PMID: 33872693 DOI: 10.1016/j.addr.2021.04.013] [Citation(s) in RCA: 112] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 03/20/2021] [Accepted: 04/15/2021] [Indexed: 12/13/2022]
Abstract
Extracellular vesicles (EVs) are biological nanoparticles naturally secreted by cells, acting as delivery vehicles for molecular messages. During the last decade, EVs have been assigned multiple functions that have established their potential as therapeutic mediators for a variety of diseases and conditions. In this review paper, we report on the potential of EVs in tissue repair and regeneration. The regenerative properties that have been associated with EVs are explored, detailing the molecular cargo they carry that is capable of mediating such effects, the signaling cascades triggered in target cells and the functional outcome achieved. EV interactions and biodistribution in vivo that influence their regenerative effects are also described, particularly upon administration in combination with biomaterials. Finally, we review the progress that has been made for the successful implementation of EV regenerative therapies in a clinical setting.
Collapse
Affiliation(s)
- Anika Nagelkerke
- Pharmaceutical Analysis, Groningen Research Institute of Pharmacy, University of Groningen, P.O. Box 196, XB20, 9700 AD Groningen, the Netherlands.
| | - Miina Ojansivu
- Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden.
| | - Luke van der Koog
- Molecular Pharmacology, Groningen Research Institute of Pharmacy, University of Groningen, P.O. Box 196, XB10, 9700 AD Groningen, the Netherlands; GRIAC Research Institute, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands.
| | - Thomas E Whittaker
- Department of Materials, Imperial College London, London, UK; Department of Bioengineering, Imperial College London, London, UK; Institute of Biomedical Engineering, Imperial College London, London, UK
| | - Eoghan M Cunnane
- Department of Materials, Imperial College London, London, UK; Department of Bioengineering, Imperial College London, London, UK; Institute of Biomedical Engineering, Imperial College London, London, UK.
| | - Andreia M Silva
- Discovery Biology, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden.
| | - Niek Dekker
- Discovery Biology, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden.
| | - Molly M Stevens
- Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden; Department of Materials, Imperial College London, London, UK; Department of Bioengineering, Imperial College London, London, UK; Institute of Biomedical Engineering, Imperial College London, London, UK.
| |
Collapse
|
44
|
Lee JY, Kim HS. Extracellular Vesicles in Regenerative Medicine: Potentials and Challenges. Tissue Eng Regen Med 2021; 18:479-484. [PMID: 34297340 PMCID: PMC8300067 DOI: 10.1007/s13770-021-00365-w] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 05/21/2021] [Accepted: 06/14/2021] [Indexed: 12/25/2022] Open
Abstract
The ultimate goal of regenerative medicine is to regain or restore the damaged or lost function of tissues and organs. Several therapeutic strategies are currently being explored to achieve this goal. From the point of view of regenerative medicine, extracellular vesicles (EVs) are exceptionally attractive due to the fact that they can overcome the limitations faced by many cell therapies and can be engineered according to their purpose through various technical modifications. EVs are biological nanoscale vesicles naturally secreted by all forms of living organisms, including prokaryotes and eukaryotes, and act as vehicles of communication between cells and their surrounding environment. Over the past decade, EVs have emerged as a new therapeutic agent for various diseases and conditions owing to their multifaceted biological functions. This is reflected by the number of publications on this subject found in the Web of Science database, which currently exceeds 12,300, over 85% of which were published within the last decade, demonstrating the increasing global trends of this innovative field. The reviews collected in this special issue provide an overview of the different approaches being explored in the use of EVs for regenerative medicine.
Collapse
Affiliation(s)
- Ji Yong Lee
- Research Institute of Hyperbaric Medicine and Science, Yonsei University Wonju College of Medicine, Wonju-si, Gangwon-do, 26426, Republic of Korea
| | - Han-Soo Kim
- Department of Biomedical Sciences, Catholic Kwandong University College of Medical Convergence, Gangneung-si, Gangwon-do, 25601, Republic of Korea.
| |
Collapse
|
45
|
Zhang X, Jiang Y, Huang Q, Wu Z, Pu H, Xu Z, Li B, Lu X, Yang X, Qin J, Peng Z. Exosomes derived from adipose-derived stem cells overexpressing glyoxalase-1 protect endothelial cells and enhance angiogenesis in type 2 diabetic mice with limb ischemia. Stem Cell Res Ther 2021; 12:403. [PMID: 34266474 PMCID: PMC8281719 DOI: 10.1186/s13287-021-02475-7] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 06/26/2021] [Indexed: 01/09/2023] Open
Abstract
Background Diabetic limb ischemia is a clinical syndrome and refractory to therapy. Our previous study demonstrated that adipose-derived stem cells (ADSCs) overexpressing glyoxalase-1 (GLO-1) promoted the regeneration of ischemic lower limbs in diabetic mice, but low survival rate, difficulty in differentiation, and tumorigenicity of the transplanted cells restricted its application. Recent studies have found that exosomes secreted by the ADSCs have the advantages of containing parental beneficial factors and exhibiting non-immunogenic, non-tumorigenic, and strong stable characteristics. Methods ADSCs overexpressing GLO-1 (G-ADSCs) were established using lentivirus transfection, and exosomes secreted from ADSCs (G-ADSC-Exos) were isolated and characterized to coculture with human umbilical vein endothelial cells (HUVECs). Proliferation, apoptosis, migration, and tube formation of the HUVECs were detected under high-glucose conditions. The G-ADSC-Exos were injected into ischemic hindlimb muscles of type 2 diabetes mellitus (T2DM) mice, and the laser Doppler perfusion index, Masson’s staining, immunofluorescence, and immunohistochemistry assays were adopted to assess the treatment efficiency. Moreover, the underlying regulatory mechanisms of the G-ADSC-Exos on the proliferation, migration, angiogenesis, and apoptosis of the HUVECs were explored. Results The G-ADSC-Exos enhanced the proliferation, migration, tube formation, and anti-apoptosis of the HUVECs in vitro under high-glucose conditions. After in vivo transplantation, the G-ADSC-Exo group showed significantly higher laser Doppler perfusion index, better muscle structural integrity, and higher microvessel’s density than the ADSC-Exo and control groups by Masson’s staining and immunofluorescence assays. The underlying mechanisms by which the G-ADSC-Exos protected endothelial cells both in vitro and in vivo might be via the activation of eNOS/AKT/ERK/P-38 signaling pathways, inhibition of AP-1/ROS/NLRP3/ASC/Caspase-1/IL-1β, as well as the increased secretion of VEGF, IGF-1, and FGF. Conclusion Exosomes derived from adipose-derived stem cells overexpressing GLO-1 protected the endothelial cells and promoted the angiogenesis in type 2 diabetic mice with limb ischemia, which will be a promising clinical treatment in diabetic lower limb ischemia. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-021-02475-7.
Collapse
Affiliation(s)
- Xing Zhang
- Department of Vascular Surgery, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200011, China
| | - Yihong Jiang
- Department of Vascular Surgery, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200011, China
| | - Qun Huang
- Department of Vascular Surgery, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200011, China
| | - Zhaoyu Wu
- Department of Vascular Surgery, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200011, China
| | - Hongji Pu
- Department of Vascular Surgery, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200011, China
| | - Zhijue Xu
- Department of Vascular Surgery, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200011, China
| | - Bo Li
- Department of Vascular Surgery, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200011, China
| | - Xinwu Lu
- Department of Vascular Surgery, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200011, China
| | - Xinrui Yang
- Department of Vascular Surgery, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200011, China.
| | - Jinbao Qin
- Department of Vascular Surgery, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200011, China.
| | - Zhiyou Peng
- Department of Vascular Surgery, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200011, China.
| |
Collapse
|
46
|
Efimenko AY, Kalinina NI, Rubina KA, Semina EV, Sysoeva VY, Akopyan ZA, Tkachuk VA. Secretome of Multipotent Mesenchymal Stromal Cells as a Promising Treatment and for Rehabilitation of Patients with the Novel Coronaviral Infection. HERALD OF THE RUSSIAN ACADEMY OF SCIENCES 2021; 91:170-175. [PMID: 34131372 PMCID: PMC8192105 DOI: 10.1134/s101933162102012x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/25/2020] [Revised: 12/28/2020] [Accepted: 01/30/2021] [Indexed: 06/12/2023]
Abstract
As a rule, coronavirus infections are mild in healthy adults and do not require special approaches to treatment. However, highly pathogenic strains, particularly the recently isolated SARS-CoV2, which causes COVID-19 infection, in about 15% of cases lead to severe complications, including acute respiratory distress syndrome, which causes high patient mortality. In addition, a common complication of COVID-19 is the development of pulmonary fibrosis. Why is the novel coronavirus so pathogenic? What new treatments can be proposed to speed up the recovery and subsequent rehabilitation of the organism? In 2020, over 34 000 scientific articles were published on the structure, distribution, pathogenesis, and possible approaches to the treatment of infection caused by the novel SARS-CoV2 coronavirus. However, there are still no definitive answers to these questions, while the number of the diseased is increasing daily. One of the comprehensive approaches to the treatment of the consequences of the infection is the use of multipotent human mesenchymal stromal cells and products of their secretion (secretome). Acting at several stages of the development of the infection, the components of the secretome can suppress the interaction of the virus with endothelial cells, regulate inflammation, and stimulate lung tissue regeneration, preventing the development of fibrosis. The results of basic and clinical research on this topic are summarized, including our own experimental data, indicating that cell therapy approaches can be successfully applied to treat patients with COVID-19.
Collapse
Affiliation(s)
- A. Yu. Efimenko
- Moscow State University, Moscow, Russia
- Institute for Regenerative Medicine, Medical Research and Educational Center, Moscow State University, Moscow, Russia
| | | | | | - E. V. Semina
- Moscow State University, Moscow, Russia
- National Medical Research Center of Cardiology, Ministry of Health of Russia, Moscow, Russia
| | | | - Zh. A. Akopyan
- Moscow State University, Moscow, Russia
- Institute for Regenerative Medicine, Medical Research and Educational Center, Moscow State University, Moscow, Russia
| | - V. A. Tkachuk
- Moscow State University, Moscow, Russia
- Institute for Regenerative Medicine, Medical Research and Educational Center, Moscow State University, Moscow, Russia
| |
Collapse
|
47
|
Zheng C, Sui B, Zhang X, Hu J, Chen J, Liu J, Wu D, Ye Q, Xiang L, Qiu X, Liu S, Deng Z, Zhou J, Liu S, Shi S, Jin Y. Apoptotic vesicles restore liver macrophage homeostasis to counteract type 2 diabetes. J Extracell Vesicles 2021; 10:e12109. [PMID: 34084287 PMCID: PMC8144839 DOI: 10.1002/jev2.12109] [Citation(s) in RCA: 129] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 04/26/2021] [Accepted: 05/12/2021] [Indexed: 12/12/2022] Open
Abstract
Apoptosis is a naturally occurring process generating plenty of apoptotic vesicles (apoVs), but the feature, fate and function of apoVs remain largely unknown. Notably, as an appealing source for cell therapy, mesenchymal stem cells (MSCs) undergo necessary apoptosis and release apoVs during therapeutic application. In this study, we characterized and used MSC‐derived apoVs to treat type 2 diabetes (T2D) mice, and we found that apoVs were efferocytosed by macrophages and functionally modulated liver macrophage homeostasis to counteract T2D. We showed that apoVs can induce macrophage reprogramming at the transcription level in an efferocytosis‐dependent manner, leading to inhibition of macrophage accumulation and transformation of macrophages towards an anti‐inflammation phenotype in T2D liver. At the molecular level, we discovered that calreticulin (CRT) was exposed on the surface of apoVs to act as a critical ‘eat‐me’ signal mediating apoV efferocytosis and macrophage regulatory effects. Importantly, we demonstrated that CRT‐mediated efferocytosis of MSC‐derived apoVs contributes to T2D therapy with alleviation of T2D phenotypes including glucose intolerance and insulin resistance. These findings uncover that functional efferocytosis of apoVs restores liver macrophage homeostasis and ameliorates T2D.
Collapse
Affiliation(s)
- Chenxi Zheng
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases Center for Tissue Engineering School of Stomatology The Fourth Military Medical University Xi'an Shaanxi China
| | - Bingdong Sui
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases Center for Tissue Engineering School of Stomatology The Fourth Military Medical University Xi'an Shaanxi China.,Department of Anatomy and Cell Biology School of Dental Medicine University of Pennsylvania Philadelphia Pennsylvania USA
| | - Xiao Zhang
- Department of Prosthodontics National Laboratory for Digital and Material Technology of Stomatology Beijing Key Laboratory of Digital Stomatology National Clinical Research Center for Oral Diseases Peking University School and Hospital of Stomatology Beijing China.,Department of Anatomy and Cell Biology School of Dental Medicine University of Pennsylvania Philadelphia Pennsylvania USA
| | - Jiachen Hu
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases Center for Tissue Engineering School of Stomatology The Fourth Military Medical University Xi'an Shaanxi China
| | - Ji Chen
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases Center for Tissue Engineering School of Stomatology The Fourth Military Medical University Xi'an Shaanxi China
| | - Jin Liu
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases Center for Tissue Engineering School of Stomatology The Fourth Military Medical University Xi'an Shaanxi China
| | - Di Wu
- South China Center of Craniofacial Stem Cell Research Guanghua School and Hospital of Stomatology Sun Yat-sen University Guangzhou Guangdong China
| | - Qingyuan Ye
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases Center for Tissue Engineering School of Stomatology The Fourth Military Medical University Xi'an Shaanxi China
| | - Lei Xiang
- South China Center of Craniofacial Stem Cell Research Guanghua School and Hospital of Stomatology Sun Yat-sen University Guangzhou Guangdong China
| | - Xinyu Qiu
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases Center for Tissue Engineering School of Stomatology The Fourth Military Medical University Xi'an Shaanxi China
| | - Siying Liu
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases Center for Tissue Engineering School of Stomatology The Fourth Military Medical University Xi'an Shaanxi China
| | - Zhihong Deng
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases Center for Tissue Engineering School of Stomatology The Fourth Military Medical University Xi'an Shaanxi China
| | - Jun Zhou
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases Center for Tissue Engineering School of Stomatology The Fourth Military Medical University Xi'an Shaanxi China
| | - Shiyu Liu
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases Center for Tissue Engineering School of Stomatology The Fourth Military Medical University Xi'an Shaanxi China
| | - Songtao Shi
- South China Center of Craniofacial Stem Cell Research Guanghua School and Hospital of Stomatology Sun Yat-sen University Guangzhou Guangdong China.,Department of Anatomy and Cell Biology School of Dental Medicine University of Pennsylvania Philadelphia Pennsylvania USA
| | - Yan Jin
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases Center for Tissue Engineering School of Stomatology The Fourth Military Medical University Xi'an Shaanxi China
| |
Collapse
|
48
|
Kim H, Lee JW, Han G, Kim K, Yang Y, Kim SH. Extracellular Vesicles as Potential Theranostic Platforms for Skin Diseases and Aging. Pharmaceutics 2021; 13:pharmaceutics13050760. [PMID: 34065468 PMCID: PMC8161370 DOI: 10.3390/pharmaceutics13050760] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 05/11/2021] [Accepted: 05/17/2021] [Indexed: 02/06/2023] Open
Abstract
Extracellular vesicles (EVs), naturally secreted by cells, act as mediators for communication between cells. They are transported to the recipient cells along with cargoes such as nucleic acids, proteins, and lipids that reflect the changes occurring within the parent cells. Thus, EVs have been recognized as potential theranostic agents for diagnosis, treatment, and prognosis. In particular, the evidence accumulated to date suggests an important role of EVs in the initiation and progression of skin aging and various skin diseases, including psoriasis, systemic lupus erythematosus, vitiligo, and chronic wounds. This review highlights recent research that investigates the role of EVs and their potential as biomarkers and therapeutic agents for skin diseases and aging.
Collapse
Affiliation(s)
- Hyosuk Kim
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea
| | - Jong Won Lee
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Korea
| | - Geonhee Han
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Korea
| | - Kwangmeyung Kim
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Korea
| | - Yoosoo Yang
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea
| | - Sun Hwa Kim
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea
| |
Collapse
|
49
|
Exosomes from Adipose Mesenchymal Stem Cells Overexpressing Stanniocalcin-1 Promote Reendothelialization After Carotid Endarterium Mechanical Injury. Stem Cell Rev Rep 2021; 18:1041-1053. [PMID: 33982245 DOI: 10.1007/s12015-021-10180-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/03/2021] [Indexed: 02/07/2023]
Abstract
OBJECTIVE Stanniocalcin-1 (STC-1) is a secreted glycoprotein that participates in the regulation of inflammation, apoptosis, and necrosis. We investigated the reendothelialization effect of exosomes from adipose stem cells (ADSC) overexpressing STC-1 on injured carotid endarterium. METHODS ADSCs were transfected with lentivirus vectors containing pre-STC-1. PHK-26 as molecular probe was used to track the exosomes engulfed by mice arterial endothelial cells (MAEC). The role of STC-1-ADSC-Exosome (S-ADSC-Exo) in MAECs was verified through scratch test and tube forming. Expressions of STC-1 and NLRP3 inflammasome were detected by western blot and quantitative reverse transcription polymerase chain reaction. Reendothelialization effect was inhibited by the antagonist of siRNA targeting STC-1. Carotid endarterium mechanical injury was induced by insertion with a guidewire into the common carotid artery lumen. Carotid arteries were harvested for histological examination, immunofluorescence staining, and Evan's blue staining. RESULTS Transfection of STC-1 significantly enhanced STC-1 levels in ADSCs, their exosomes, and MAECs. Compared with the control group and the ADSC-Exo group, STC-1 enriched exosomes markedly inhibited the expressions of NLRP3, Caspase-1, and IL-1β in MAECs, exhibited good lateral migration capacity, and promoted angiogenesis. Administration of siRNA targeting STC-1 completely abolished down-regulation of NLRP3, Caspase-1, and IL-1β by STC-1 and inhibited effects of S-ADSC-Exo on lateral migration and angiogenesis. In vivo administration of S-ADSC-Exo had reendothelialization effect on post-injury carotid endarterium as evidenced by thinner arterial wall, low-expressed NLRP3 inflammasome, and more living endothelial cells. CONCLUSIONS The reendothelialization effect of exosomes from ADSCs on post-injury carotid endarterium could be enhanced by genetic modification of the exosomes to contain elevated STC-1, possibly through suppression of NLRP3 inflammasome-mediated inflammation.
Collapse
|
50
|
Extracellular Vesicles from Human Adipose-Derived Mesenchymal Stem Cells: A Review of Common Cargos. Stem Cell Rev Rep 2021; 18:854-901. [PMID: 33904115 PMCID: PMC8942954 DOI: 10.1007/s12015-021-10155-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/14/2021] [Indexed: 12/14/2022]
Abstract
In recent years, the interest in adipose tissue mesenchymal cell–derived extracellular vesicles (AT-MSC-EVs) has increasingly grown. Numerous articles support the potential of human AT-MSC-EVs as a new therapeutic option for treatment of diverse diseases in the musculoskeletal and cardiovascular systems, kidney, skin, and immune system, among others. This approach makes use of the molecules transported inside of EVs, which play an important role in cell communication and in transmission of macromolecules. However, to our knowledge, there is no database where essential information about AT-MSC-EVs cargo molecules is gathered for easy reference. The aim of this study is to describe the different molecules reported so far in AT-MSC- EVs, their main molecular functions, and biological processes in which they are involved. Recently, the presence of 591 proteins and 604 microRNAs (miRNAs) has been described in human AT-MSC-EVs. The main molecular function enabled by both proteins and miRNAs present in human AT-MSC-EVs is the binding function. Signal transduction and gene silencing are the biological processes in which a greater number of proteins and miRNAs from human AT-MSC-EVs are involved, respectively. In this review we highlight the therapeutics effects of AT-MSC-EVs related with their participation in relevant biological processes including inflammation, angiogenesis, cell proliferation, apoptosis and migration, among others.
Collapse
|