1
|
Kou Y, Jin Z, Yuan Y, Ma B, Xie W, Han N. FK506 contributes to peripheral nerve regeneration by inhibiting neuroinflammatory responses and promoting neuron survival. Neural Regen Res 2025; 20:2108-2115. [PMID: 39254569 PMCID: PMC11691453 DOI: 10.4103/nrr.nrr-d-22-00867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 11/23/2023] [Accepted: 01/17/2024] [Indexed: 09/11/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202507000-00031/figure1/v/2024-09-09T124005Z/r/image-tiff FK506 (Tacrolimus) is a systemic immunosuppressant approved by the U.S. Food and Drug Administration. FK506 has been shown to promote peripheral nerve regeneration, however, its precise mechanism of action and its pathways remain unclear. In this study, we established a rat model of sciatic nerve injury and found that FK506 improved the morphology of the injured sciatic nerve, increased the numbers of motor and sensory neurons, reduced inflammatory responses, markedly improved the conduction function of the injured nerve, and promoted motor function recovery. These findings suggest that FK506 promotes peripheral nerve structure recovery and functional regeneration by reducing the intensity of inflammation after neuronal injury and increasing the number of surviving neurons.
Collapse
Affiliation(s)
- Yuhui Kou
- Department of Trauma and Orthopedics, Peking University People’s Hospital, Beijing, China
- National Center for Trauma Medicine, Peking University People’s Hospital, Beijing, China
- Key Laboratory of Trauma and Neural Regeneration (Peking University), Ministry of Education, Beijing, China
| | - Zongxue Jin
- National Center for Trauma Medicine, Peking University People’s Hospital, Beijing, China
- Trauma Medicine Center, Peking University People’s Hospital, Beijing, China
| | - Yusong Yuan
- Department of Orthopedics, China-Japan Friendship Hospital, Beijing, China
| | - Bo Ma
- Department of Trauma and Orthopedics, Peking University People’s Hospital, Beijing, China
- Key Laboratory of Trauma and Neural Regeneration (Peking University), Ministry of Education, Beijing, China
| | - Wenyong Xie
- Department of Trauma and Orthopedics, Peking University People’s Hospital, Beijing, China
| | - Na Han
- National Center for Trauma Medicine, Peking University People’s Hospital, Beijing, China
- Key Laboratory of Trauma and Neural Regeneration (Peking University), Ministry of Education, Beijing, China
| |
Collapse
|
2
|
Chen D, Chen Z, Yuan J, Chen G, Chen Y, He K, Hu Y, Ye L, Yang Y. Research landscape and trends of human umbilical cord mesenchymal stem cell-derived exosomes. Stem Cell Res Ther 2025; 16:259. [PMID: 40437553 PMCID: PMC12121053 DOI: 10.1186/s13287-025-04379-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Accepted: 05/02/2025] [Indexed: 06/01/2025] Open
Abstract
BACKGROUND Human umbilical cord mesenchymal stem cell-derived exosomes (hUCMSC-Exos) have gained significant attention for their potential in cellular regeneration and functional rehabilitation. Nevertheless, the rapid expansion of research in this field makes it challenging for emerging trends and strategic priorities, potentially impeding scientific advancement. This study employs bibliometric analysis to systematically evaluate the research landscape and highlight pivotal research trajectories of hUCMSC-Exos. METHODS Publications on hUCMSC-Exos from 2012 to 2024 were retrieved from the Web of Science Core Collection (WoSCC). Quantitative bibliometric analysis was implemented through integrated utilization of VOSviewer, CiteSpace, and Bibliometrix analytical tools. RESULTS China and its institutions led global publication output, with Qian Hui from Jiangsu University identified as the most prolific author. STEM CELL RESEARCH & THERAPY emerged as a high-impact journal in this domain. Current research predominantly focuses on immunomodulation, regenerative medicine, pharmaceutical delivery systems, and clinical model development. Future research directions are expected to explore angiogenesis, spinal cord injury, and immunomodulation. CONCLUSIONS This study maps the evolving landscape of hUCMSC-Exos research, emphasizing its applications in regenerative medicine. By synthesizing current and emerging paradigms, these findings provide insights into therapeutic potential, novel mechanisms, and pathways for clinical translation.
Collapse
Affiliation(s)
- Desheng Chen
- Department of Hepatic Surgery and Liver Transplantation Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Guangdong Key Laboratory of Liver Disease Research, Key Laboratory of Liver Disease Biotherapy and Translational Medicine of Guangdong Higher Education Institutes, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Zeping Chen
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiabin Yuan
- Department of Orthopedics, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Guanzi Chen
- Department of Infectious Diseases, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yutao Chen
- Department of Hepatic Surgery and Liver Transplantation Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Guangdong Key Laboratory of Liver Disease Research, Key Laboratory of Liver Disease Biotherapy and Translational Medicine of Guangdong Higher Education Institutes, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Kaiming He
- Department of Hepatic Surgery and Liver Transplantation Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Guangdong Key Laboratory of Liver Disease Research, Key Laboratory of Liver Disease Biotherapy and Translational Medicine of Guangdong Higher Education Institutes, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yongwei Hu
- Department of Hepatic Surgery and Liver Transplantation Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.
- Guangdong Key Laboratory of Liver Disease Research, Key Laboratory of Liver Disease Biotherapy and Translational Medicine of Guangdong Higher Education Institutes, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.
| | - Linsen Ye
- Department of Hepatic Surgery and Liver Transplantation Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.
- Guangdong Key Laboratory of Liver Disease Research, Key Laboratory of Liver Disease Biotherapy and Translational Medicine of Guangdong Higher Education Institutes, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.
| | - Yang Yang
- Department of Hepatic Surgery and Liver Transplantation Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.
- Guangdong Key Laboratory of Liver Disease Research, Key Laboratory of Liver Disease Biotherapy and Translational Medicine of Guangdong Higher Education Institutes, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
3
|
Li C, Meng X, Li S, Wang C. Therapeutic Advances in Peripheral Nerve Injuries: Nerve-Guided Conduit and Beyond. TISSUE ENGINEERING. PART B, REVIEWS 2025. [PMID: 40195945 DOI: 10.1089/ten.teb.2024.0322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Peripheral nerve injury (PNI), a challenging neurosurgery issue, often leads to partial or complete loss of neuronal functions and even neuropathic pain. Thus far, the gold standard for treating peripheral nerve deficit remains autografts. While numerous reviews have explored PNI and regeneration, this work distinctively synthesizes recent advancements in tissue engineering-particularly four-dimensional (4D) bioprinting and exosome therapies-with an emphasis on their clinical translation. By consolidating findings spanning molecular mechanisms to therapeutic applications, this review proposes an actionable framework for advancing experimental strategies toward clinically viable solutions. Our work critically evaluates emerging innovations such as dynamically adaptive 4D-printed nerve conduits and exosome-based therapies, underscoring their potential to match conventional autografts in achieving functional restoration. Impact Statement Although several previous reviews have been made on describing with great detail the degenerative and regenerative mechanisms of the peripheral nervous systems, as well as the several existing and exploratory treatment strategies, we focus more on the latest advancements of each of those topics.
Collapse
Affiliation(s)
- Changqing Li
- First Clinical Medical College, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Xianyu Meng
- The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| | - Shengji Li
- First Clinical Medical College, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Chengjing Wang
- First Clinical Medical College, Heilongjiang University of Chinese Medicine, Harbin, China
| |
Collapse
|
4
|
Nevado-Sánchez E, Rodríguez-Díaz M, Núñez-Rodríguez S, Bueno-de la Fuente A, de la Fuente-Anuncibay R, Villar-Suárez V, González-Bernal JJ, Labrador J. Effectiveness of Stem Cell Secretomes in the Regeneration and Functional Recovery of Severed Nerves in Patients with Nerve Injuries: A Systematic Review. Cells 2025; 14:492. [PMID: 40214446 PMCID: PMC11987723 DOI: 10.3390/cells14070492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 03/13/2025] [Accepted: 03/21/2025] [Indexed: 04/14/2025] Open
Abstract
The regenerative potential of mesenchymal stem cell (MSC) secretomes in peripheral nerve injuries warrants rigorous evaluation. This systematic review analyzes their effectiveness in preclinical models of neurotmesis, a complete transection of a nerve. Neurophysiological recovery was assessed through nerve conduction velocity (NCV), a measure of the speed at which electrical impulses travel along a nerve. Following PRISMA guidelines, a systematic search was conducted in PubMed, Scopus, Web of Science, and ScienceDirect (last search July 2024). From 640 initially identified studies, 13 met inclusion criteria, encompassing 514 animals (rats). experimental designs published since 2014 in English or Spanish, focusing on MSC secretomes for nerve regeneration. Exclusion criteria included reviews, case reports, and incomplete data. The risk of bias was assessed using Joanna Briggs Institute tools. Results were synthesized narratively, focusing on functional and structural outcomes. The included studies employed various MSC sources, including adipose tissue, olfactory mucosa, and umbilical cord. Nine studies reported enhanced SFI, favoring secretome-treated groups over controls (mean difference +20.5%, p < 0.01). Seven studies documented increased NCV, with up to 35% higher conduction velocities in treated groups (p < 0.05). Histological outcomes reported in 12 studies showed increased axonal diameter (+25%, p < 0.01), myelin sheath thickness (+30%, p < 0.05), and Schwann cell proliferation. Limitations of the included evidence include methodological heterogeneity and variability in outcome measurement tools. MSC-derived secretomes demonstrate potential as advanced therapeutic strategies for nerve injuries. Personalized approaches considering injury type and clinical context are essential for optimizing outcomes.
Collapse
Affiliation(s)
- Endika Nevado-Sánchez
- Reconstructive and Aesthetic Plastic Surgery Service, Hospital Universitario de Burgos, 09006 Burgos, Spain;
| | | | | | | | | | - Vega Villar-Suárez
- Department of Surgery, Medicine and Veterinary Anatomy, Institute of Biomedicine (IBIOMED), Faculty of Veterinary Sciences, Campus de Vegazana, University of León, León 24071, Spain;
| | | | - Jorge Labrador
- Research Unit, Hospital Universitario de Burgos, 09006 Burgos, Spain;
| |
Collapse
|
5
|
Wang YJ, Chen ZH, Shen YT, Wang KX, Han YM, Zhang C, Yang XM, Chen BQ. Stem cell therapy: A promising therapeutic approach for skeletal muscle atrophy. World J Stem Cells 2025; 17:98693. [DOI: 10.4252/wjsc.v17.i2.98693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 12/09/2024] [Accepted: 01/23/2025] [Indexed: 02/24/2025] Open
Abstract
Skeletal muscle atrophy results from disruptions in the growth and metabolism of striated muscle, leading to a reduction or loss of muscle fibers. This condition not only significantly impacts patients’ quality of life but also imposes substantial socioeconomic burdens. The complex molecular mechanisms driving skeletal muscle atrophy contribute to the absence of effective treatment options. Recent advances in stem cell therapy have positioned it as a promising approach for addressing this condition. This article reviews the molecular mechanisms of muscle atrophy and outlines current therapeutic strategies, focusing on mesenchymal stem cells, induced pluripotent stem cells, and their derivatives. Additionally, the challenges these stem cells face in clinical applications are discussed. A deeper understanding of the regenerative potential of various stem cells could pave the way for breakthroughs in the prevention and treatment of muscle atrophy.
Collapse
Affiliation(s)
- Ying-Jie Wang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Nantong University, Nantong 226000, Jiangsu Province, China
| | - Ze-Hao Chen
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Nantong University, Nantong 226000, Jiangsu Province, China
| | - Yun-Tian Shen
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Nantong University, Nantong 226000, Jiangsu Province, China
| | - Ke-Xin Wang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Nantong University, Nantong 226000, Jiangsu Province, China
| | - Yi-Min Han
- Medical College, Nantong University, Nantong 226000, Jiangsu Province, China
| | - Chen Zhang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Nantong University, Nantong 226000, Jiangsu Province, China
| | - Xiao-Ming Yang
- Co-Innovation Center of Neuroregeneration, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Nantong University, Nantong 226000, Jiangsu Province, China
- Research and Development Center for E-Learning, Ministry of Education, Beijing 100816, China
| | - Bing-Qian Chen
- Department of Orthopaedics, Changshu Hospital Affiliated to Soochow University, Changshu 215500, Jiangsu Province, China
| |
Collapse
|
6
|
Thakur A, Rai D. Global requirements for manufacturing and validation of clinical grade extracellular vesicles. THE JOURNAL OF LIQUID BIOPSY 2024; 6:100278. [PMID: 40027307 PMCID: PMC11863704 DOI: 10.1016/j.jlb.2024.100278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 11/17/2024] [Accepted: 11/18/2024] [Indexed: 03/05/2025]
Abstract
Extracellular vesicles (EVs) are nanovesicles released from different cell types from biofluids such as blood, urine, and cerebrospinal fluid. They vary in size and biomarkers, and their biogenesis pathways allow them to be divided into three major types: exosomes, micro-vesicles, and apoptotic bodies. EVs have been studied in the context of diagnosis and therapeutic intervention of various pathological conditions such as cancer, neurodegenerative diseases, and pulmonary diseases. However, the production of EV-based therapeutics can be affected by the source, heterogeneity, or disease, raising questions about the manufacturing and validation of EVs of clinical grade and their scope regarding good manufacturing practice (GMP) in the industry. To address this, we have discussed the state-of-the-art requirements for EV production that must occur in a GMP-compliant environment with a reliable and traceable source. Additionally, EVs' homogeneity and the therapeutics' purity and stability must be analyzed and validated. Quality control measures must also be established to ensure the safety and efficacy of EVs. In conclusion, these considerations must be weighed carefully when manufacturing and validating EVs of clinical grade to ensure their safety and efficacy for therapeutic use.
Collapse
Affiliation(s)
- Abhimanyu Thakur
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Deepika Rai
- Smidt Heart Institute, Cedars-Sinai Medical Centre, Los Angeles, CA, United States
| |
Collapse
|
7
|
Liu M, Chen X. Human Umbilical Cord-Derived Mesenchymal Stem Cells-Exosomes-Delivered miR-375 Targets HDAC4 to Promote Autophagy and Suppress T Cell Apoptosis in Sepsis-Associated Acute Kidney Injury. Appl Biochem Biotechnol 2024; 196:7954-7973. [PMID: 38668845 DOI: 10.1007/s12010-024-04963-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/16/2024] [Indexed: 12/14/2024]
Abstract
This study sought to elucidate the mechanism of human umbilical cord-derived mesenchymal stem cells (HUCMSCs)-exosomes (Exos) in sepsis-associated acute kidney injury (SAKI). Exos were isolated from HUCMSCs and co-cultured with CD4+ T cells exposed to lipopolysaccharide to detect the effects of HUCMSCs-Exos on CD4+ T cell apoptosis and autophagy. miR-375 expression in CD4+ T cells and HUCMSCs-Exos was examined. The relationship between miR-375 and HDAC4 was analyzed. A mouse model of SAKI was established and injected with HUCMSCs-Exos to verify the function of HUCMSCs-Exos in vivo. HUCMSCs-Exos inhibited lipopolysaccharide-induced apoptosis of CD4+ T cells and promoted autophagy. miR-375 expression was noted to be elevated in the HUCMSCs-Exos. Importantly, HUCMSCs-Exos could deliver miR-375 into CD4+ T cells where miR-375 targeted HDAC4 and negatively regulated its expression. By this mechanism, HUCMSCs-Exos decreased CD4+ T cell apoptosis and augmented autophagy. This finding was further confirmed in an in vivo SAKI model. Collectively, HUCMSCs-Exos can protect against SAKI via delivering miR-375 that promotes autophagy and arrests T cell apoptosis through HDAC4 downregulation. These findings suggest a promising therapeutic potential for HUCMSCs-Exos in the context of SAKI.
Collapse
Affiliation(s)
- Min Liu
- Department of Intensive Care, the First Hospital of Changsha, No. 311 Yingpan Road, Changsha, Hunan, 410005, People's Republic of China
| | - Xiyun Chen
- Department of Gynecology, the First Hospital of Changsha, No. 311 Yingpan Road, Changsha, Hunan, 410005, People's Republic of China.
| |
Collapse
|
8
|
Pedersen C, Chen VT, Herbst P, Zhang R, Elfert A, Krishan A, Azar DT, Chang JH, Hu WY, Kremsmayer TP, Jalilian E, Djalilian AR, Guaiquil VH, Rosenblatt MI. Target specification and therapeutic potential of extracellular vesicles for regulating corneal angiogenesis, lymphangiogenesis, and nerve repair. Ocul Surf 2024; 34:459-476. [PMID: 39426677 PMCID: PMC11921040 DOI: 10.1016/j.jtos.2024.10.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 09/16/2024] [Accepted: 10/15/2024] [Indexed: 10/21/2024]
Abstract
Extracellular vesicles, including exosomes, are small extracellular vesicles that range in size from 30 nm to 10 μm in diameter and have specific membrane markers. They are naturally secreted and are present in various bodily fluids, including blood, urine, and saliva, and through the variety of their internal cargo, they contribute to both normal physiological and pathological processes. These processes include immune modulation, neuronal synapse formation, cell differentiation, cancer metastasis, angiogenesis, lymphangiogenesis, progression of infectious disease, and neurodegenerative disorders like Alzheimer's and Parkinson's disease. In recent years, interest has grown in the use of exosomes as a potential drug delivery system for various diseases and injuries. Importantly, exosomes originating from a patient's own cells exhibit minimal immunogenicity and possess remarkable stability along with inherent and adjustable targeting capabilities. This review explores the roles of exosomes in angiogenesis, lymphangiogenesis, and nerve repair with a specific emphasis on these processes within the cornea. Furthermore, it examines exosomes derived from specific cell types, discusses the advantages of exosome-based therapies in modulating these processes, and presents some of the most established methods for exosome isolation. Exosome-based treatments are emerging as potential minimally invasive and non-immunogenic therapies that modulate corneal angiogenesis and lymphangiogenesis, as well as enhance and accelerate endogenous corneal nerve repair.
Collapse
Affiliation(s)
- Cameron Pedersen
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Victoria T Chen
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Paula Herbst
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Runze Zhang
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Amr Elfert
- University of Illinois Cancer Center, Chicago, IL, USA
| | - Abhi Krishan
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Dimitri T Azar
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Jin-Hong Chang
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA; Jesse Brown Veterans Affairs Medical Center, Chicago, IL, USA.
| | - Wen-Yang Hu
- Department of Urology, University of Illinois at Chicago, Chicago, IL, USA
| | - Tobias P Kremsmayer
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Elmira Jalilian
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA; Richard and Loan Hill Department of Bioengineering, University of Illinois at Chicago, Chicago, IL, 60607, USA
| | - Ali R Djalilian
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Victor H Guaiquil
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Mark I Rosenblatt
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA.
| |
Collapse
|
9
|
Putthanbut N, Lee JY, Borlongan CV. Extracellular vesicle therapy in neurological disorders. J Biomed Sci 2024; 31:85. [PMID: 39183263 PMCID: PMC11346291 DOI: 10.1186/s12929-024-01075-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 08/16/2024] [Indexed: 08/27/2024] Open
Abstract
Extracellular vesicles (EVs) are vital for cell-to-cell communication, transferring proteins, lipids, and nucleic acids in various physiological and pathological processes. They play crucial roles in immune modulation and tissue regeneration but are also involved in pathogenic conditions like inflammation and degenerative disorders. EVs have heterogeneous populations and cargo, with numerous subpopulations currently under investigations. EV therapy shows promise in stimulating tissue repair and serving as a drug delivery vehicle, offering advantages over cell therapy, such as ease of engineering and minimal risk of tumorigenesis. However, challenges remain, including inconsistent nomenclature, complex characterization, and underdeveloped large-scale production protocols. This review highlights the recent advances and significance of EVs heterogeneity, emphasizing the need for a better understanding of their roles in disease pathologies to develop tailored EV therapies for clinical applications in neurological disorders.
Collapse
Affiliation(s)
- Napasiri Putthanbut
- Department of Neurosurgery, Center of Aging and Brain Repair, University of South Florida, Tampa, USA
- Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Salaya, Thailand
| | - Jea Young Lee
- Department of Neurosurgery, Center of Aging and Brain Repair, University of South Florida, Tampa, USA
| | - Cesario V Borlongan
- Department of Neurosurgery, Center of Aging and Brain Repair, University of South Florida, Tampa, USA.
| |
Collapse
|
10
|
Rasouli A, Roshangar L, Hosseini M, Pourmohammadfazel A, Nikzad S. Beyond boundaries: The therapeutic potential of exosomes in neural microenvironments in neurological disorders. Neuroscience 2024; 553:98-109. [PMID: 38964450 DOI: 10.1016/j.neuroscience.2024.06.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 06/18/2024] [Accepted: 06/25/2024] [Indexed: 07/06/2024]
Abstract
Neurological disorders are a diverse group of conditions that can significantly impact individuals' quality of life. The maintenance of neural microenvironment homeostasis is essential for optimal physiological cellular processes. Perturbations in this delicate balance underlie various pathological manifestations observed across various neurological disorders. Current treatments for neurological disorders face substantial challenges, primarily due to the formidable blood-brain barrier and the intricate nature of neural tissue structures. These obstacles have resulted in a paucity of effective therapies and inefficiencies in patient care. Exosomes, nanoscale vesicles that contain a complex repertoire of biomolecules, are identifiable in various bodily fluids. They hold substantial promise in numerous therapeutic interventions due to their unique attributes, including targeted drug delivery mechanisms and the ability to cross the BBB, thereby enhancing their therapeutic potential. In this review, we investigate the therapeutic potential of exosomes across a range of neurological disorders, including neurodegenerative disorders, traumatic brain injury, peripheral nerve injury, brain tumors, and stroke. Through both in vitro and in vivo studies, our findings underscore the beneficial influence of exosomes in enhancing the neural microenvironment following neurological diseases, offering promise for improved neural recovery and management in these conditions.
Collapse
Affiliation(s)
- Arefe Rasouli
- Department of Anatomical Sciences, School of Medicine Tabriz University of Medical Sciences, Tabriz, Iran
| | - Leila Roshangar
- Department of Anatomical Sciences, School of Medicine Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Mohammadbagher Hosseini
- Department of Pediatrics, School of Medicine Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Pourmohammadfazel
- Department of Anatomical Sciences, School of Medicine Tabriz University of Medical Sciences, Tabriz, Iran
| | | |
Collapse
|
11
|
Sharifi M, Kamalabadi-Farahani M, Salehi M, Ebrahimi-Brough S, Alizadeh M. Recent perspectives on the synergy of mesenchymal stem cells with micro/nano strategies in peripheral nerve regeneration-a review. Front Bioeng Biotechnol 2024; 12:1401512. [PMID: 39050683 PMCID: PMC11266111 DOI: 10.3389/fbioe.2024.1401512] [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: 03/15/2024] [Accepted: 06/19/2024] [Indexed: 07/27/2024] Open
Abstract
Despite the intrinsic repair of peripheral nerve injury (PNI), it is important to carefully monitor the process of peripheral nerve repair, as peripheral nerve regeneration is slow and incomplete in large traumatic lesions. Hence, mesenchymal stem cells (MSCs) with protective and regenerative functions are utilized in synergy with innovative micro/nano technologies to enhance the regeneration process of peripheral nerves. Nonetheless, as MSCs are assessed using standard regenerative criteria including sensory-motor indices, structural features, and morphology, it is challenging to differentiate between the protective and regenerative impacts of MSCs on neural tissue. This study aims to analyze the process of nerve regeneration, particularly the performance of MSCs with and without synergistic approaches. It also focuses on the paracrine secretions of MSCs and their conversion into neurons with functional properties that influence nerve regeneration after PNI. Furthermore, the study explores new ideas for nerve regeneration after PNI by considering the synergistic effect of MSCs and therapeutic compounds, neuronal cell derivatives, biological or polymeric conduits, organic/inorganic nanoparticles, and electrical stimulation. Finally, the study highlights the main obstacles to developing synergy in nerve regeneration after PNI and aims to open new windows based on recent advances in neural tissue regeneration.
Collapse
Affiliation(s)
- Majid Sharifi
- Student Research Committee, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
- Tissue Engineering and Stem Cells Research Center, Shahroud University of Medical Sciences, Shahroud, Iran
- Department of Tissue Engineering, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Mohammad Kamalabadi-Farahani
- Tissue Engineering and Stem Cells Research Center, Shahroud University of Medical Sciences, Shahroud, Iran
- Department of Tissue Engineering, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Majid Salehi
- Tissue Engineering and Stem Cells Research Center, Shahroud University of Medical Sciences, Shahroud, Iran
- Department of Tissue Engineering, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
- Health Technology Incubator Center, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Somayeh Ebrahimi-Brough
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Morteza Alizadeh
- Department of Tissue Engineering and Biomaterials, School of Advanced Medical Sciences and Technologies, Hamadan University of Medical Sciences, Hamadan, Iran
| |
Collapse
|
12
|
Fang N, Wang Z, Jiang J, Yang A, Mao T, Wang Z, Chen Q. Nonsurgical therapy for lumbar spinal stenosis caused by ligamentum flavum hypertrophy: A review. Medicine (Baltimore) 2024; 103:e38782. [PMID: 38968524 PMCID: PMC11224896 DOI: 10.1097/md.0000000000038782] [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: 05/15/2024] [Accepted: 06/11/2024] [Indexed: 07/07/2024] Open
Abstract
Lumbar spinal stenosis (LSS) can cause a range of cauda equina symptoms, including lower back and leg pain, numbness, and intermittent claudication. This disease affects approximately 103 million people worldwide, particularly the elderly, and can seriously compromise their health and well-being. Ligamentum flavum hypertrophy (LFH) is one of the main contributing factors to this disease. Surgical treatment is currently recommended for LSS caused by LFH. For patients who do not meet the criteria for surgery, symptom relief can be achieved by using oral nonsteroidal anti-inflammatory drugs (NSAIDs) and epidural steroid injections. Exercise therapy and needle knife can also help to reduce the effects of mechanical stress. However, the effectiveness of these methods varies, and targeting the delay in LF hypertrophy is challenging. Therefore, further research and development of new drugs is necessary to address this issue. Several new drugs, including cyclopamine and N-acetyl-l-cysteine, are currently undergoing testing and may serve as new treatments for LSS caused by LFH.
Collapse
Affiliation(s)
- Nan Fang
- College of Acupuncture & Orthopedics, Hubei University of Chinese Medicine, Wuhan, China
- Department of Orthopedics & Traumatology, Hubei Provincial Hospital of TCM, Wuhan, China
| | - Zhigang Wang
- Department of Orthopedics & Traumatology, Hubei Provincial Hospital of TCM, Wuhan, China
- Department of Orthopedics & Traumatology, Affiliated Hospital of Hubei University of Chinese Medicine, Wuhan, China
| | - Jiecheng Jiang
- College of Acupuncture & Orthopedics, Hubei University of Chinese Medicine, Wuhan, China
- Department of Orthopedics & Traumatology, Hubei Provincial Hospital of TCM, Wuhan, China
| | - Aofei Yang
- Department of Orthopedics & Traumatology, Hubei Provincial Hospital of TCM, Wuhan, China
- Department of Orthopedics & Traumatology, Affiliated Hospital of Hubei University of Chinese Medicine, Wuhan, China
| | - Tian Mao
- Department of Orthopedics & Traumatology, Hubei Provincial Hospital of TCM, Wuhan, China
- Department of Orthopedics & Traumatology, Affiliated Hospital of Hubei University of Chinese Medicine, Wuhan, China
| | - Zitong Wang
- College of Acupuncture & Orthopedics, Hubei University of Chinese Medicine, Wuhan, China
- Department of Orthopedics & Traumatology, Hubei Provincial Hospital of TCM, Wuhan, China
| | - Qian Chen
- College of Acupuncture & Orthopedics, Hubei University of Chinese Medicine, Wuhan, China
- Department of Orthopedics & Traumatology, Hubei Provincial Hospital of TCM, Wuhan, China
| |
Collapse
|
13
|
Bahar R, Darabi S, Norouzian M, Roustaei S, Torkamani-Dordshaikh S, Hasanzadeh M, Vakili K, Fathi M, Khodagholi F, Kaveh N, Jahanbaz S, Moghaddam MH, Abbaszadeh HA, Aliaghaei A. Neuroprotective effect of human cord blood-derived extracellular vesicles by improved neuromuscular function and reduced gliosis in a rat model of Huntington's disease. J Chem Neuroanat 2024; 138:102419. [PMID: 38609056 DOI: 10.1016/j.jchemneu.2024.102419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 04/09/2024] [Accepted: 04/09/2024] [Indexed: 04/14/2024]
Abstract
Huntington's disease (HD) is a hereditary condition characterized by the gradual deterioration of nerve cells in the striatum. Recent scientific investigations have revealed the promising potential of Extracellular vesicles (EVs) as a therapy to mitigate inflammation and enhance motor function. This study aimed to examine the impact of administering EVs derived from human umbilical cord blood (HUCB) on the motor abilities and inflammation levels in a rat model of HD. After ultracentrifugation to prepare EVs from HUCB to determine the nature of the obtained contents, the expression of CD markers 81 and 9, the average size and also the morphology of its particles were investigated by DLS and Transmission electron microscopy (TEM). Then, in order to induce the HD model, 3-nitropropionic acid (3-NP) neurotoxin was injected intraperitoneal into the rats, after treatment by HUCB-EVs, rotarod, electromyogram (EMG) and the open field tests were performed on the rats. Finally, after rat sacrifice and the striatum was removed, Hematoxylin and eosin staining (H&E), stereology, immunohistochemistry, antioxidant tests, and western blot were performed. Our results showed that the contents of the HUCB-EVs express the CD9 and CD81 markers and have spherical shapes. In addition, the injection of HUCB-EVs improved motor and neuromuscular function, reduced gliosis, increased antioxidant activity and inflammatory factor, and partially prevented the decrease of neurons. The findings generally show that HUCB-EVs have neuroprotective effects and reduce neuroinflammation from the toxic effects of 3-NP, which can be beneficial for the recovery of HD.
Collapse
Affiliation(s)
- Reza Bahar
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Laser Application in Medical Sciences Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Shahram Darabi
- Cellular and Molecular Research Center, Research Institute for Non-Communicable Diseases, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Mohsen Norouzian
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Susan Roustaei
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Hearing Disorders Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Shayesteh Torkamani-Dordshaikh
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Laser Application in Medical Sciences Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Maral Hasanzadeh
- Hearing Disorders Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Kimia Vakili
- Laser Application in Medical Sciences Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mobina Fathi
- Laser Application in Medical Sciences Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fariba Khodagholi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Neda Kaveh
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Shima Jahanbaz
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Laser Application in Medical Sciences Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Meysam Hassani Moghaddam
- Department of Anatomical Sciences, Faculty of Medicine, AJA University of Medical Sciences, Tehran, Iran
| | - Hojjat-Allah Abbaszadeh
- Laser Application in Medical Sciences Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Abbas Aliaghaei
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
14
|
Yavuz B, Mutlu EC, Ahmed Z, Ben-Nissan B, Stamboulis A. Applications of Stem Cell-Derived Extracellular Vesicles in Nerve Regeneration. Int J Mol Sci 2024; 25:5863. [PMID: 38892052 PMCID: PMC11172915 DOI: 10.3390/ijms25115863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 05/15/2024] [Accepted: 05/23/2024] [Indexed: 06/21/2024] Open
Abstract
Extracellular vesicles (EVs), including exosomes, microvesicles, and other lipid vesicles derived from cells, play a pivotal role in intercellular communication by transferring information between cells. EVs secreted by progenitor and stem cells have been associated with the therapeutic effects observed in cell-based therapies, and they also contribute to tissue regeneration following injury, such as in orthopaedic surgery cases. This review explores the involvement of EVs in nerve regeneration, their potential as drug carriers, and their significance in stem cell research and cell-free therapies. It underscores the importance of bioengineers comprehending and manipulating EV activity to optimize the efficacy of tissue engineering and regenerative therapies.
Collapse
Affiliation(s)
- Burcak Yavuz
- Vocational School of Health Services, Altinbas University, 34147 Istanbul, Turkey;
| | - Esra Cansever Mutlu
- Biomaterials Research Group, School of Metallurgy and Materials, College of Engineering and Physical Science, University of Birmingham, Birmingham B15 2TT, UK;
| | - Zubair Ahmed
- Neuroscience & Ophthalmology, Institute of Inflammation and Ageing, University of Birmingham, Edgbaston B15 2TT, UK
| | - Besim Ben-Nissan
- Translational Biomaterials and Medicine Group, School of Life Sciences, University of Technology Sydney, P.O. Box 123, Broadway, NSW 2007, Australia;
| | - Artemis Stamboulis
- Biomaterials Research Group, School of Metallurgy and Materials, College of Engineering and Physical Science, University of Birmingham, Birmingham B15 2TT, UK;
| |
Collapse
|
15
|
Li S, Zhang J, Liu X, Wang N, Sun L, Liu J, Liu X, Masoudi A, Wang H, Li C, Guo C, Liu X. Proteomic characterization of hUC-MSC extracellular vesicles and evaluation of its therapeutic potential to treat Alzheimer's disease. Sci Rep 2024; 14:5959. [PMID: 38472335 PMCID: PMC10933327 DOI: 10.1038/s41598-024-56549-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 03/07/2024] [Indexed: 03/14/2024] Open
Abstract
In recent years, human umbilical cord mesenchymal stem cell (hUC-MSC) extracellular vesicles (EVs) have been used as a cell replacement therapy and have been shown to effectively overcome some of the disadvantages of cell therapy. However, the specific mechanism of action of EVs is still unclear, and there is no appropriate system for characterizing the differences in the molecular active substances of EVs produced by cells in different physiological states. We used a data-independent acquisition (DIA) quantitative proteomics method to identify and quantify the protein composition of two generations EVs from three different donors and analysed the function and possible mechanism of action of the proteins in EVs of hUC-MSCs via bioinformatics. By comparative proteomic analysis, we characterized the different passages EVs. Furthermore, we found that adaptor-related protein complex 2 subunit alpha 1 (AP2A1) and adaptor-related protein complex 2 subunit beta 1 (AP2B1) in hUC-MSC-derived EVs may play a significant role in the treatment of Alzheimer's disease (AD) by regulating the synaptic vesicle cycle signalling pathway. Our work provides a direction for batch-to-batch quality control of hUC-MSC-derived EVs and their application in AD treatment.
Collapse
Affiliation(s)
- Shuang Li
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Hebei Anti-Tumour Molecular Target Technology Innovation Center, College of Life Science, Hebei Normal University, Shijiazhuang, 050024, China
| | - Jiayi Zhang
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Hebei Anti-Tumour Molecular Target Technology Innovation Center, College of Life Science, Hebei Normal University, Shijiazhuang, 050024, China
| | - Xinxing Liu
- Jianyuan Precision Medicines (Zhangjiakou) Co., Ltd., Zhangjiakou, 075000, China
| | - Ningmei Wang
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Hebei Anti-Tumour Molecular Target Technology Innovation Center, College of Life Science, Hebei Normal University, Shijiazhuang, 050024, China
| | - Luyao Sun
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Hebei Anti-Tumour Molecular Target Technology Innovation Center, College of Life Science, Hebei Normal University, Shijiazhuang, 050024, China
| | - Jianling Liu
- Jianyuan Precision Medicines (Zhangjiakou) Co., Ltd., Zhangjiakou, 075000, China
- Cancer Research Institute, The First Affiliated Hospital of Hebei North University, Zhangjiakou, 075000, China
| | - Xingliang Liu
- Department of Neurology, The First Affiliated Hospital of Hebei North University, Zhangjiakou, 075000, China
| | - Abolfazl Masoudi
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Hebei Anti-Tumour Molecular Target Technology Innovation Center, College of Life Science, Hebei Normal University, Shijiazhuang, 050024, China
| | - Hui Wang
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Hebei Anti-Tumour Molecular Target Technology Innovation Center, College of Life Science, Hebei Normal University, Shijiazhuang, 050024, China
| | - Chunxia Li
- Obstetrics and Gynaecology, The Fifth Hospital of Zhangjiakou, Zhangjiakou, 075000, China
| | - Chunyan Guo
- Hebei Key Laboratory of Neuropharmacology; Department of Pharmacy, Hebei North University, Zhangjiakou, 075000, China.
| | - Xifu Liu
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Hebei Anti-Tumour Molecular Target Technology Innovation Center, College of Life Science, Hebei Normal University, Shijiazhuang, 050024, China.
| |
Collapse
|
16
|
Goryunov K, Ivanov M, Kulikov A, Shevtsova Y, Burov A, Podurovskaya Y, Zubkov V, Degtyarev D, Sukhikh G, Silachev D. A Review of the Use of Extracellular Vesicles in the Treatment of Neonatal Diseases: Current State and Problems with Translation to the Clinic. Int J Mol Sci 2024; 25:2879. [PMID: 38474125 PMCID: PMC10932115 DOI: 10.3390/ijms25052879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 02/22/2024] [Accepted: 02/27/2024] [Indexed: 03/14/2024] Open
Abstract
Neonatal disorders, particularly those resulting from prematurity, pose a major challenge in health care and have a significant impact on infant mortality and long-term child health. The limitations of current therapeutic strategies emphasize the need for innovative treatments. New cell-free technologies utilizing extracellular vesicles (EVs) offer a compelling opportunity for neonatal therapy by harnessing the inherent regenerative capabilities of EVs. These nanoscale particles, secreted by a variety of organisms including animals, bacteria, fungi and plants, contain a repertoire of bioactive molecules with therapeutic potential. This review aims to provide a comprehensive assessment of the therapeutic effects of EVs and mechanistic insights into EVs from stem cells, biological fluids and non-animal sources, with a focus on common neonatal conditions such as hypoxic-ischemic encephalopathy, respiratory distress syndrome, bronchopulmonary dysplasia and necrotizing enterocolitis. This review summarizes evidence for the therapeutic potential of EVs, analyzes evidence of their mechanisms of action and discusses the challenges associated with the implementation of EV-based therapies in neonatal clinical practice.
Collapse
Affiliation(s)
- Kirill Goryunov
- V.I. Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Moscow 117198, Russia; (K.G.); (M.I.); (Y.S.); (A.B.); (Y.P.); (V.Z.); (D.D.); (G.S.)
| | - Mikhail Ivanov
- V.I. Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Moscow 117198, Russia; (K.G.); (M.I.); (Y.S.); (A.B.); (Y.P.); (V.Z.); (D.D.); (G.S.)
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119992, Russia
| | - Andrey Kulikov
- Medical Institute, Patrice Lumumba Peoples’ Friendship University of Russia (RUDN University), Moscow 117198, Russia;
| | - Yulia Shevtsova
- V.I. Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Moscow 117198, Russia; (K.G.); (M.I.); (Y.S.); (A.B.); (Y.P.); (V.Z.); (D.D.); (G.S.)
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119992, Russia
| | - Artem Burov
- V.I. Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Moscow 117198, Russia; (K.G.); (M.I.); (Y.S.); (A.B.); (Y.P.); (V.Z.); (D.D.); (G.S.)
| | - Yulia Podurovskaya
- V.I. Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Moscow 117198, Russia; (K.G.); (M.I.); (Y.S.); (A.B.); (Y.P.); (V.Z.); (D.D.); (G.S.)
| | - Victor Zubkov
- V.I. Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Moscow 117198, Russia; (K.G.); (M.I.); (Y.S.); (A.B.); (Y.P.); (V.Z.); (D.D.); (G.S.)
| | - Dmitry Degtyarev
- V.I. Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Moscow 117198, Russia; (K.G.); (M.I.); (Y.S.); (A.B.); (Y.P.); (V.Z.); (D.D.); (G.S.)
| | - Gennady Sukhikh
- V.I. Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Moscow 117198, Russia; (K.G.); (M.I.); (Y.S.); (A.B.); (Y.P.); (V.Z.); (D.D.); (G.S.)
| | - Denis Silachev
- V.I. Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Moscow 117198, Russia; (K.G.); (M.I.); (Y.S.); (A.B.); (Y.P.); (V.Z.); (D.D.); (G.S.)
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119992, Russia
| |
Collapse
|
17
|
Zhang Y, Yi D, Hong Q, Cao J, Geng X, Liu J, Xu C, Cao M, Chen C, Xu S, Zhang Z, Li M, Zhu Y, Peng N. Platelet-rich plasma-derived exosomes boost mesenchymal stem cells to promote peripheral nerve regeneration. J Control Release 2024; 367:265-282. [PMID: 38253204 DOI: 10.1016/j.jconrel.2024.01.043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 01/10/2024] [Accepted: 01/19/2024] [Indexed: 01/24/2024]
Abstract
Peripheral nerve injury (PNI) remains a severe clinical problem with debilitating consequences. Mesenchymal stem cell (MSC)-based therapy is promising, but the problems of poor engraftment and insufficient neurotrophic effects need to be overcome. Herein, we isolated platelet-rich plasma-derived exosomes (PRP-Exos), which contain abundant bioactive molecules, and investigated their potential to increase the regenerative capacity of MSCs. We observed that PRP-Exos significantly increased MSC proliferation, viability, and mobility, decreased MSC apoptosis under stress, maintained MSC stemness, and attenuated MSC senescence. In vivo, PRP-Exo-treated MSCs (pExo-MSCs) exhibited an increased retention rate and heightened therapeutic efficacy, as indicated by increased axonal regeneration, remyelination, and recovery of neurological function in a PNI model. In vitro, pExo-MSCs coculture promoted Schwann cell proliferation and dorsal root ganglion axon growth. Moreover, the increased neurotrophic behaviour of pExo-MSCs was mediated by trophic factors, particularly glia-derived neurotrophic factor (GDNF), and PRP-Exos activated the PI3K/Akt signalling pathway in MSCs, leading to the observed phenotypes. These findings demonstrate that PRP-Exos may be novel agents for increasing the ability of MSCs to promote neural repair and regeneration in patients with PNI.
Collapse
Affiliation(s)
- Yongyi Zhang
- Medical School of Chinese PLA, Beijing 100853, China; Department of Rehabilitation Medicine, The Second Medical Centre & National Clinical Research Centre for Geriatric Diseases, Chinese PLA General Hospital, Beijing 100853, China; State Key Laboratory of Kidney Diseases, Nephrology Institute of the Chinese PLA, National Clinical Research Centre for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, The First Medical Centre, Chinese PLA General Hospital, Beijing 100853, China; No.962 Hospital of the PLA Joint Logistic Support Force, Harbin 150080, China
| | - Dan Yi
- Medical School of Chinese PLA, Beijing 100853, China; Departments of Ultrasound, The First Medical Centre, Chinese PLA General Hospital, Beijing 100853, China
| | - Quan Hong
- State Key Laboratory of Kidney Diseases, Nephrology Institute of the Chinese PLA, National Clinical Research Centre for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, The First Medical Centre, Chinese PLA General Hospital, Beijing 100853, China
| | - Jiangbei Cao
- Departments of Anaesthesiology, The First Medical Centre, Chinese PLA General Hospital, Beijing 100853, China
| | - Xiaodong Geng
- State Key Laboratory of Kidney Diseases, Nephrology Institute of the Chinese PLA, National Clinical Research Centre for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, The First Medical Centre, Chinese PLA General Hospital, Beijing 100853, China
| | - Jinwei Liu
- Medical School of Chinese PLA, Beijing 100853, China; Department of Rehabilitation Medicine, The Second Medical Centre & National Clinical Research Centre for Geriatric Diseases, Chinese PLA General Hospital, Beijing 100853, China
| | - Chuang Xu
- Medical School of Chinese PLA, Beijing 100853, China; Department of Rehabilitation Medicine, The Second Medical Centre & National Clinical Research Centre for Geriatric Diseases, Chinese PLA General Hospital, Beijing 100853, China
| | - Mengyu Cao
- Department of Rehabilitation Medicine, The Second Medical Centre & National Clinical Research Centre for Geriatric Diseases, Chinese PLA General Hospital, Beijing 100853, China
| | - Chao Chen
- Medical School of Chinese PLA, Beijing 100853, China; Department of Rehabilitation Medicine, The Second Medical Centre & National Clinical Research Centre for Geriatric Diseases, Chinese PLA General Hospital, Beijing 100853, China
| | - Shuaixuan Xu
- Medical School of Chinese PLA, Beijing 100853, China; Department of Rehabilitation Medicine, The Second Medical Centre & National Clinical Research Centre for Geriatric Diseases, Chinese PLA General Hospital, Beijing 100853, China
| | - Zhen Zhang
- Medical School of Chinese PLA, Beijing 100853, China; Department of Rehabilitation Medicine, The Second Medical Centre & National Clinical Research Centre for Geriatric Diseases, Chinese PLA General Hospital, Beijing 100853, China
| | - Molin Li
- Medical School of Chinese PLA, Beijing 100853, China; Departments of Ultrasound, The First Medical Centre, Chinese PLA General Hospital, Beijing 100853, China
| | - Yaqiong Zhu
- Departments of Ultrasound, The First Medical Centre, Chinese PLA General Hospital, Beijing 100853, China.
| | - Nan Peng
- Medical School of Chinese PLA, Beijing 100853, China; Department of Rehabilitation Medicine, The Second Medical Centre & National Clinical Research Centre for Geriatric Diseases, Chinese PLA General Hospital, Beijing 100853, China.
| |
Collapse
|
18
|
Zhao X, Deng H, Feng Y, Wang Y, Yao X, Ma Y, Zhang L, Jie J, Yang P, Yang Y. Immune-cell-mediated tissue engineering strategies for peripheral nerve injury and regeneration. J Mater Chem B 2024; 12:2217-2235. [PMID: 38345580 DOI: 10.1039/d3tb02557h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/29/2024]
Abstract
During the process of peripheral nerve repair, there are many complex pathological and physiological changes, including multi-cellular responses and various signaling molecules, and all these events establish a dynamic microenvironment for axon repair, regeneration, and target tissue/organ reinnervation. The immune system plays an indispensable role in the process of nerve repair and function recovery. An effective immune response not only involves innate-immune and adaptive-immune cells but also consists of chemokines and cytokines released by these immune cells. The elucidation of the orchestrated interplay of immune cells with nerve regeneration and functional restoration is meaningful for the exploration of therapeutic strategies. This review mainly enumerates the general immune cell response to peripheral nerve injury and focuses on their contributions to functional recovery. The tissue engineering-mediated strategies to regulate macrophages and T cells through physical and biochemical factors combined with scaffolds are discussed. The dynamic immune responses during peripheral nerve repair and immune-cell-mediated tissue engineering methods are presented, which provide a new insight and inspiration for immunomodulatory therapies in peripheral nerve regeneration.
Collapse
Affiliation(s)
- Xueying Zhao
- Key Laboratory of Neuroregeneration of Jiangsu Province and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, 226001, Nantong, P. R. China.
| | - Hui Deng
- Key Laboratory of Neuroregeneration of Jiangsu Province and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, 226001, Nantong, P. R. China.
| | - Yuan Feng
- Key Laboratory of Neuroregeneration of Jiangsu Province and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, 226001, Nantong, P. R. China.
| | - Yuehan Wang
- Key Laboratory of Neuroregeneration of Jiangsu Province and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, 226001, Nantong, P. R. China.
| | - Xiaomin Yao
- Key Laboratory of Neuroregeneration of Jiangsu Province and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, 226001, Nantong, P. R. China.
| | - Yuyang Ma
- Key Laboratory of Neuroregeneration of Jiangsu Province and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, 226001, Nantong, P. R. China.
| | - Luzhong Zhang
- Key Laboratory of Neuroregeneration of Jiangsu Province and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, 226001, Nantong, P. R. China.
| | - Jing Jie
- Department of Clinical Laboratory, The Second Affiliated Hospital of Nantong University, 226001, Nantong, P. R. China.
| | - Pengxiang Yang
- Key Laboratory of Neuroregeneration of Jiangsu Province and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, 226001, Nantong, P. R. China.
| | - Yumin Yang
- Key Laboratory of Neuroregeneration of Jiangsu Province and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, 226001, Nantong, P. R. China.
| |
Collapse
|
19
|
Min XL, Liu HJ, Dou XK, Chen FX, Zhao Q, Zhao XH, Shi Y, Zhao QY, Sun SJ, Wang Z, Yu SH. Extracellular Vesicles from Neural Stem Cells Carry microRNA-16-5p to Reduce Corticosterone-induced Neuronal Injury in Depression Rats. Neuroscience 2024; 538:95-109. [PMID: 37778691 DOI: 10.1016/j.neuroscience.2023.09.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 09/19/2023] [Accepted: 09/25/2023] [Indexed: 10/03/2023]
Abstract
OBJECTIVE Depression is a common mental illness. Neural stem cell-derived extracellular vesicles (NSC-EVs) are involved in repairing neuronal injury. We estimated the mechanism of miR-16-5p in depression rats. METHODS EVs were extracted from NSCs. The depression rat model was established by corticosterone (CORT) induction and treated with NSC-EVs. The depression behavioral/pathological changes in rats were assessed using forced swimming test, open field test, sucrose consumption test and western blotting. The neuronal apoptosis in hippocampal tissue were detected. CORT-induced PC12 cell model was established. EV uptake by PC12 cells was measured and PC12 cell apoptosis was detected. The downstream targets of miR-16-5p were predicted and verified. The expressions of miR-16-5p and MYB in rats, PC12 cells, and EVs were measured. Functional rescue experiments were conducted to verify the role of miR-16-5p and MYB in PC12 cell apoptosis. RESULTS CORT induction increased neuronal apoptosis in hippocampal tissue and induced depression-like behaviors in rats, while NSC-EV treatment improved depression-like behaviors and apoptosis in rats. In PC12 cells, NSC-EVs decreased CORT-induced PC12 cell apoptosis. NSC-EVs carried miR-16-5p into PC12 cells. miR-16-5p knockdown in EVs partially reversed the inhibitory effects of NSC-EVs on CORT-induced PC12 cell apoptosis. miR-16-5p targeted to inhibit MYB to repress CORT-induced PC12 cell apoptosis. In vivo experiments further verified that NSC-EVs reduced neuronal injury in CORT-induced depression rats via the miR-16-5p/MYB axis. CONCLUSION NSC-EVs-mediated alleviation on neuronal injury by carrying miR-16-5p to target MYB was highly likely one of the mechanisms by which NSC-EVs mediated miR-16-5p in neuroprotection of depression rats.
Collapse
Affiliation(s)
- Xiao-Li Min
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Kunming Medical University, Kunming, China.
| | - Hai-Jing Liu
- Department of Acupuncture and Massage, Yunnan Traditional Chinese Medicine University, Kunming, China
| | - Xing-Kui Dou
- Department of Cardiology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Fei-Xiong Chen
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Qing Zhao
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Xiao-Hong Zhao
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Ying Shi
- Department of Internal Medicine, Clinical Medicine School, Yunnan Traditional Chinese Medicine University, Kunming, China
| | - Qun-Yuan Zhao
- Department of Emergency, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Sheng-Jie Sun
- Department of Emergency, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Zhen Wang
- Department of Neurosurgery, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Si-Hang Yu
- Department of Neurosurgery, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| |
Collapse
|
20
|
Morishita M, Kida M, Motomura T, Tsukamoto R, Atari M, Higashiwaki K, Masuda K, Katsumi H, Yamamoto A. Elucidation of the Tissue Distribution and Host Immunostimulatory Activity of Exogenously Administered Probiotic-Derived Extracellular Vesicles for Immunoadjuvant. Mol Pharm 2023; 20:6104-6113. [PMID: 37931251 DOI: 10.1021/acs.molpharmaceut.3c00460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2023]
Abstract
Extracellular vesicles (EVs) are cell-derived nanoparticles that can be used as novel biomaterials. In the development of EVs-based therapeutic systems, it is essential to understand the in vivo fate of exogenously administered EVs and subsequent biological responses mediated by EVs. Although probiotics and microorganisms that modulate the host immune system also secrete EVs, their tissue distribution and biological reactions after administration to the host have not been sufficiently elucidated. In this study, we characterized EVs released from the probiotics Bifidobacterium longum (B-EVs) and Lactobacillus plantarum WCFS1 (L-EVs) in terms of tissue distribution and immune-activating capacity after intravenous and subcutaneous administration in mice. B-EVs and L-EVs exhibited particle sizes of approximately 100-160 nm and negative zeta potentials. These EVs contained peptidoglycan, DNA, and RNA as their cargoes. Intravenously administered B-EVs and L-EVs mainly accumulated in the liver and spleen. Furthermore, liver F4/80 and splenic CD169 macrophages took up the intravenously administered EVs. Subcutaneously administered B-EVs and L-EVs accumulated in the lymph nodes and were mainly located in the B-lymphocyte zone, indicating that exogenously administered probiotic-derived EVs showed a similar biodistribution, irrespective of the EVs-secreting cell type. Evaluation of EVs-mediated immune reactions demonstrated that intravenously administered EVs showed little activation potency. In contrast, subcutaneously administered B-EVs strongly increased the expression of inflammatory cytokine (TNF-α) and co-stimulatory molecules (CD40 and CD80) than L-EVs. These findings indicate that the subcutaneous administration of B-EVs is a useful strategy for the development of novel EVs-based immunotherapies.
Collapse
Affiliation(s)
- Masaki Morishita
- Department of Biopharmaceutics, Kyoto Pharmaceutical University, Misasagi, Yamashina-Ku, Kyoto 607-8414, Japan
| | - Masakatsu Kida
- Department of Biopharmaceutics, Kyoto Pharmaceutical University, Misasagi, Yamashina-Ku, Kyoto 607-8414, Japan
| | - Tomomi Motomura
- Department of Biopharmaceutics, Kyoto Pharmaceutical University, Misasagi, Yamashina-Ku, Kyoto 607-8414, Japan
| | - Rihito Tsukamoto
- Department of Biopharmaceutics, Kyoto Pharmaceutical University, Misasagi, Yamashina-Ku, Kyoto 607-8414, Japan
| | - Mizuho Atari
- Department of Biopharmaceutics, Kyoto Pharmaceutical University, Misasagi, Yamashina-Ku, Kyoto 607-8414, Japan
| | - Kazuya Higashiwaki
- Department of Biopharmaceutics, Kyoto Pharmaceutical University, Misasagi, Yamashina-Ku, Kyoto 607-8414, Japan
| | - Kisa Masuda
- Department of Biopharmaceutics, Kyoto Pharmaceutical University, Misasagi, Yamashina-Ku, Kyoto 607-8414, Japan
| | - Hidemasa Katsumi
- Department of Biopharmaceutics, Kyoto Pharmaceutical University, Misasagi, Yamashina-Ku, Kyoto 607-8414, Japan
| | - Akira Yamamoto
- Department of Biopharmaceutics, Kyoto Pharmaceutical University, Misasagi, Yamashina-Ku, Kyoto 607-8414, Japan
| |
Collapse
|
21
|
Yi D, Zhang Y, Li M, Chen J, Chen X, Wang L, Xing G, Chen S, Zhu Y, Wang Y. Ultrasound-Targeted Microbubble Destruction Assisted Delivery of Platelet-Rich Plasma-Derived Exosomes Promoting Peripheral Nerve Regeneration. Tissue Eng Part A 2023; 29:645-662. [PMID: 37612613 DOI: 10.1089/ten.tea.2023.0133] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/25/2023] Open
Abstract
Peripheral nerve injury is prevalent and has a high disability rate in clinical settings. Current therapeutic methods have not achieved satisfactory efficacy, underscoring the need for novel approaches to nerve restoration that remains an active area of research in neuroscience and regenerative medicine. In this study, we isolated platelet-rich plasma-derived exosomes (PRP-exos) and found that they can significantly enhance the proliferation, migration, and secretion of trophic factors by Schwann cells (SCs). In addition, there were marked changes in transcriptional and expression profiles of SCs, particularly via the upregulation of genes related to biological functions involved in nerve regeneration and repair. In the rat model of sciatic nerve crush injury, ultrasound-targeted microbubble destruction (UTMD) enhanced the efficiency of PRP-exos delivery to the injury site. This approach ensured a high concentration of PRP-exos in the injured nerve and improved the therapeutic outcomes. In conclusion, PRP-exos may promote nerve regeneration and repair, and UTMD may increase the effectiveness of targeted PRP-exos delivery to the injured nerve and enhance the therapeutic effect.
Collapse
Affiliation(s)
- Dan Yi
- Medical School of Chinese PLA, Beijing, China
- Department of Ultrasound, The First Medical Centre, Chinese PLA General Hospital, Beijing, China
- Department of Nephrology, The First Medical Centre, Chinese PLA General Hospital, Nephrology Institute of the Chinese PLA, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing, China
| | - Yongyi Zhang
- Medical School of Chinese PLA, Beijing, China
- Department of Rehabilitation Medicine, The Second Medical Centre, Chinese PLA General Hospital, Beijing, China
- No.962 Hospital of the PLA Joint Logistic Support Force, Harbin, China
| | - Molin Li
- Medical School of Chinese PLA, Beijing, China
- Department of Ultrasound, The First Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Jian Chen
- Department of Ultrasound, The First Medical Centre, Chinese PLA General Hospital, Beijing, China
- Medical College of Nankai University, Tianjin, China
| | - Xianghui Chen
- Medical School of Chinese PLA, Beijing, China
- Department of Ultrasound, The First Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Li Wang
- Medical School of Chinese PLA, Beijing, China
- Department of Ultrasound, The Sixth Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Guanghui Xing
- Medical School of Chinese PLA, Beijing, China
- Department of Ultrasound, The Fourth Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Siming Chen
- Department of Ultrasound, The First Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Yaqiong Zhu
- Department of Ultrasound, The First Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Yuexiang Wang
- Department of Ultrasound, The First Medical Centre, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
22
|
Wu KY, Deng F, Mao XY, Zhou D, Shen WG. Ferroptosis involves in Schwann cell death in diabetic peripheral neuropathy. Open Med (Wars) 2023; 18:20230809. [PMID: 37829841 PMCID: PMC10566555 DOI: 10.1515/med-2023-0809] [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: 04/27/2023] [Revised: 08/16/2023] [Accepted: 09/01/2023] [Indexed: 10/14/2023] Open
Abstract
Accumulating evidence shows that Schwann cells' (SCs) death caused by high glucose (HG) is involved in the pathological process of diabetic peripheral neuropathy (DPN). Ferroptosis is a novel form of regulatory cell death driven by iron-dependent lipid peroxidation. However, it is not clear whether ferroptosis is involved in the death process of SCs induced by HG. The expression of ferroptosis-related indicators in the serum of DPN patients was detected by ELISA. Subsequently, using cell counting kit‑8, western blot, real-time PCR, and Ki-67 staining, we investigated the effects of HG on the ferroptosis of SCs and initially explored the underlying mechanism. The results showed that the serum levels of glutathione peroxidase 4 (GPX4) and glutathione in patients with DPN decreased, while malondialdehyde levels increased significantly. Then, we observed that erastin and HG induced ferroptosis in SCs, resulting in the decrease in cell activity and the expression level of GPX4 and SLC7A11, which could be effectively reversed by the ferroptosis inhibitor Fer-1. Mechanistically, HG induced ferroptosis in SCs by inhibiting the NRF2 signaling pathway. Our results showed that ferroptosis was involved in the death process of SCs induced by HG. Inhibition of ferroptosis in SCs might create a new avenue for the treatment of DPN.
Collapse
Affiliation(s)
- Kai-yan Wu
- Department of Cell Biology, School of Medicine of Yangzhou University, Yangzhou, Jiangsu, 225009, China
| | - Fei Deng
- Department of Cell Biology, School of Medicine of Yangzhou University, Yangzhou, Jiangsu, 225009, China
| | - Xin-yu Mao
- Department of Cell Biology, School of Medicine of Yangzhou University, Yangzhou, Jiangsu, 225009, China
| | - Dan Zhou
- Department of Central Laboratory, Jintan Hospital, Jiangsu University, 500 Avenue Jintan, Jintan, Jiangsu, 213200, China
| | - Wei-gan Shen
- Department of Cell Biology, School of Medicine of Yangzhou University, Yangzhou, Jiangsu, 225009, China
| |
Collapse
|
23
|
Khaled MM, Ibrahium AM, Abdelgalil AI, El-Saied MA, El-Bably SH. Regenerative Strategies in Treatment of Peripheral Nerve Injuries in Different Animal Models. Tissue Eng Regen Med 2023; 20:839-877. [PMID: 37572269 PMCID: PMC10519924 DOI: 10.1007/s13770-023-00559-4] [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: 03/28/2023] [Revised: 05/15/2023] [Accepted: 05/21/2023] [Indexed: 08/14/2023] Open
Abstract
BACKGROUND Peripheral nerve damage mainly resulted from traumatic or infectious causes; the main signs of a damaged nerve are the loss of sensory and/or motor functions. The injured nerve has limited regenerative capacity and is recovered by the body itself, the recovery process depends on the severity of damage to the nerve, nowadays the use of stem cells is one of the new and advanced methods for treatment of these problems. METHOD Following our review, data are collected from different databases "Google scholar, Springer, Elsevier, Egyptian Knowledge Bank, and PubMed" using different keywords such as Peripheral nerve damage, Radial Nerve, Sciatic Nerve, Animals, Nerve regeneration, and Stem cell to investigate the different methods taken in consideration for regeneration of PNI. RESULT This review contains tables illustrating all forms and types of regenerative medicine used in treatment of peripheral nerve injuries (PNI) including different types of stem cells " adipose-derived stem cells, bone marrow stem cells, Human umbilical cord stem cells, embryonic stem cells" and their effect on re-constitution and functional recovery of the damaged nerve which evaluated by physical, histological, Immuno-histochemical, biochemical evaluation, and the review illuminated the best regenerative strategies help in rapid peripheral nerve regeneration in different animal models included horse, dog, cat, sheep, monkey, pig, mice and rat. CONCLUSION Old surgical attempts such as neurorrhaphy, autogenic nerve transplantation, and Schwann cell implantation have a limited power of recovery in cases of large nerve defects. Stem cell therapy including mesenchymal stromal cells has a high potential differentiation capacity to renew and form a new nerve and also restore its function.
Collapse
Affiliation(s)
- Mona M Khaled
- Department of Anatomy and Embryology, Faculty of Veterinary Medicine, Cairo University, Giza Square, Giza, 12211, Egypt.
| | - Asmaa M Ibrahium
- Department of Anatomy and Embryology, Faculty of Veterinary Medicine, Cairo University, Giza Square, Giza, 12211, Egypt
| | - Ahmed I Abdelgalil
- Department of Surgery, Anaesthesiology and Radiology, Faculty of Veterinary Medicine, Cairo University, Giza Square, Giza, 12211, Egypt
| | - Mohamed A El-Saied
- Department of Pathology, Faculty of Veterinary of Veterinary Medicine, Cairo University, Giza Square, Giza, 12211, Egypt
| | - Samah H El-Bably
- Department of Anatomy and Embryology, Faculty of Veterinary Medicine, Cairo University, Giza Square, Giza, 12211, Egypt
| |
Collapse
|
24
|
Dong X, Dong JF, Zhang J. Roles and therapeutic potential of different extracellular vesicle subtypes on traumatic brain injury. Cell Commun Signal 2023; 21:211. [PMID: 37596642 PMCID: PMC10436659 DOI: 10.1186/s12964-023-01165-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Accepted: 05/13/2023] [Indexed: 08/20/2023] Open
Abstract
Traumatic brain injury (TBI) is a leading cause of injury-related disability and death around the world, but the clinical stratification, diagnosis, and treatment of complex TBI are limited. Due to their unique properties, extracellular vesicles (EVs) are emerging candidates for being biomarkers of traumatic brain injury as well as serving as potential therapeutic targets. However, the effects of different extracellular vesicle subtypes on the pathophysiology of traumatic brain injury are very different, or potentially even opposite. Before extracellular vesicles can be used as targets for TBI therapy, it is necessary to classify different extracellular vesicle subtypes according to their functions to clarify different strategies for EV-based TBI therapy. The purpose of this review is to discuss contradictory effects of different EV subtypes on TBI, and to propose treatment ideas based on different EV subtypes to maximize their benefits for the recovery of TBI patients. Video Abstract.
Collapse
Affiliation(s)
- Xinlong Dong
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No. 119, Nansihuan West Road, Fengtai District, Beijing, China.
- Beijing Key Laboratory of Central Nervous System Injury, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.
| | - Jing-Fei Dong
- Bloodworks Research Institute, Seattle, WA, USA
- Division of Hematology, Department of Medicine, School of Medicine, University of Washington, Seattle, WA, USA
| | - Jianning Zhang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
25
|
Zhang L, Liu J, Zhou C. Current aspects of small extracellular vesicles in pain process and relief. Biomater Res 2023; 27:78. [PMID: 37563666 PMCID: PMC10416402 DOI: 10.1186/s40824-023-00417-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 08/04/2023] [Indexed: 08/12/2023] Open
Abstract
Small extracellular vesicles (sEVs) have been identified as a noteworthy paracrine mechanism of intercellular communication in diagnosing and managing neurological disorders. Current research suggests that sEVs play a pivotal role in the pathological progression of pain, emphasizing their critical function in the pathological progression of pain in acute and chronic pain models. By facilitating the transfer of diverse molecules, such as proteins, nucleic acids, and metabolites, sEVs can modulate pain signaling transmission in both the central and peripheral nervous systems. Furthermore, the unique molecules conveyed by sEVs in pain disorders indicate their potential as diagnostic biomarkers. The application of sEVs derived from mesenchymal stem cells (MSCs) in regenerative pain medicine has emerged as a promising strategy for pain management. Moreover, modified sEVs have garnered considerable attention in the investigation of pathological processes and therapeutic interventions. This review presents a comprehensive overview of the current knowledge regarding the involvement of sEVs in pain pathogenesis and treatment. Nevertheless, additional research is imperative to facilitate their clinical implementation. Schematic diagram of sEVs in the biogenesis, signal transmission, diagnosis, and treatment of pain disorders. Small extracellular vesicles (sEVs) are secreted by multiple cells, loading with various biomolecules, such as miRNAs, transmembrane proteins, and amino acids. They selectively target other cells and regulating pain signal transmission. The composition of sEVs can serve as valuable biomarkers for pain diagnosis. In particular, mesenchymal stem cell-derived sEVs have shown promise as regenerative medicine for managing multiple pain disorders. Furthermore, by modifying the structure or contents of sEVs, they could potentially be used as a potent analgesic method.
Collapse
Affiliation(s)
- Lanyu Zhang
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Anesthesia & Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital of Sichuan University, Chengdu, 610041, Sichuan, China
| | - Jin Liu
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China
| | - Cheng Zhou
- Laboratory of Anesthesia & Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital of Sichuan University, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
26
|
Nasiry D, Khalatbary AR. Stem cell-derived extracellular vesicle-based therapy for nerve injury: A review of the molecular mechanisms. World Neurosurg X 2023; 19:100201. [PMID: 37181584 PMCID: PMC10173266 DOI: 10.1016/j.wnsx.2023.100201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 04/10/2023] [Accepted: 04/19/2023] [Indexed: 05/16/2023] Open
Abstract
Recent evidence suggests that stem cell therapy has beneficial effects on nerve damage. These beneficial effects were subsequently found to be exerted in part in a paracrine manner by the release of extracellular vesicles. Stem cell-secreted extracellular vesicles have shown great potential to reduce inflammation and apoptosis, optimize the function of Schwann cells, regulate genes related to regeneration, and improve behavioral performance after nerve damage. This review summarizes the current knowledge on the effect of stem cell-derived extracellular vesicles on neuroprotection and regeneration along with their molecular mechanisms after nerve damage.
Collapse
Affiliation(s)
- Davood Nasiry
- Amol Faculty of Paramedicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Ali Reza Khalatbary
- Cellular and Molecular Research Center, Mazandaran University of Medical Sciences, Sari, Iran
- Corresponding author.
| |
Collapse
|
27
|
Helissey C, Cavallero S, Guitard N, Théry H, Chargari C, François S. Revolutionizing Radiotoxicity Management with Mesenchymal Stem Cells and Their Derivatives: A Focus on Radiation-Induced Cystitis. Int J Mol Sci 2023; 24:ijms24109068. [PMID: 37240415 DOI: 10.3390/ijms24109068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Revised: 05/02/2023] [Accepted: 05/17/2023] [Indexed: 05/28/2023] Open
Abstract
Although radiation therapy plays a crucial role in cancer treatment, and techniques have improved continuously, irradiation induces side effects in healthy tissue. Radiation cystitis is a potential complication following the therapeutic irradiation of pelvic cancers and negatively impacts patients' quality of life (QoL). To date, no effective treatment is available, and this toxicity remains a therapeutic challenge. In recent times, stem cell-based therapy, particularly the use of mesenchymal stem cells (MSC), has gained attention in tissue repair and regeneration due to their easy accessibility and their ability to differentiate into several tissue types, modulate the immune system and secrete substances that help nearby cells grow and heal. In this review, we will summarize the pathophysiological mechanisms of radiation-induced injury to normal tissues, including radiation cystitis (RC). We will then discuss the therapeutic potential and limitations of MSCs and their derivatives, including packaged conditioned media and extracellular vesicles, in the management of radiotoxicity and RC.
Collapse
Affiliation(s)
- Carole Helissey
- Clinical Unit Research, HIA Bégin, 69 Avenu de Paris, 94160 Saint-Mandé, France
- Department of Radiation Biological Effects, French Armed Forces Biomedical Research Institute, Place Général Valérie André, 91220 Brétigny-sur-Orge, France
| | - Sophie Cavallero
- Department of Radiation Biological Effects, French Armed Forces Biomedical Research Institute, Place Général Valérie André, 91220 Brétigny-sur-Orge, France
| | - Nathalie Guitard
- Department of Radiation Biological Effects, French Armed Forces Biomedical Research Institute, Place Général Valérie André, 91220 Brétigny-sur-Orge, France
| | - Hélène Théry
- Department of Radiation Biological Effects, French Armed Forces Biomedical Research Institute, Place Général Valérie André, 91220 Brétigny-sur-Orge, France
| | - Cyrus Chargari
- Department of Radiation Biological Effects, French Armed Forces Biomedical Research Institute, Place Général Valérie André, 91220 Brétigny-sur-Orge, France
- Department of Radiation Oncology, Pitié Salpêtrière University Hospital, 47-83 Bd de l'Hôpital, 75013 Paris, France
| | - Sabine François
- Department of Radiation Biological Effects, French Armed Forces Biomedical Research Institute, Place Général Valérie André, 91220 Brétigny-sur-Orge, France
| |
Collapse
|
28
|
Wang Y, Sheng H, Cong M, Wang W, He Q, Li H, Li S, Zhang J, Chen Y, Guo S, Fang L, Pluchino S, Biskup E, Artemyev M, Chen F, Li Y, Chen J, Feng S, Wo Y. Spatio-temporally deciphering peripheral nerve regeneration in vivo after extracellular vesicle therapy under NIR-II fluorescence imaging. NANOSCALE 2023; 15:7991-8005. [PMID: 37067249 DOI: 10.1039/d3nr00795b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Extracellular vesicles (EVs) show potential as a therapeutic tool for peripheral nerve injury (PNI), promoting neurological regeneration. However, there are limited data on the in vivo spatio-temporal trafficking and biodistribution of EVs. In this study, we introduce a new non-invasive near-infrared fluorescence imaging strategy based on glucose-conjugated quantum dot (QDs-Glu) labeling to target and track EVs in a sciatic nerve injury rat model in real-time. Our results demonstrate that the injected EVs migrated from the uninjured site to the injured site of the nerve, with an increase in fluorescence signals detected from 4 to 7 days post-injection, indicating the release of contents from the EVs with therapeutic effects. Immunofluorescence and behavioral tests revealed that the EV therapy promoted nerve regeneration and functional recovery at 28 days post-injection. We also found a relationship between functional recovery and the NIR-II fluorescence intensity change pattern, providing novel evidence for the therapeutic effects of EV therapy using real-time NIR-II imaging at the live animal level. This approach initiates a new path for monitoring EVs in treating PNI under in vivo NIR-II imaging, enhancing our understanding of the efficacy of EV therapy on peripheral nerve regeneration and its mechanisms.
Collapse
Affiliation(s)
- Yueming Wang
- Department of Anatomy and Physiology, School of Medicine, Shanghai Jiao Tong University, Shanghai 200025, China.
| | - Huaixuan Sheng
- Sports Medicine Institute of Fudan University, Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China.
| | - Meng Cong
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Nantong University, Nantong, JS 226001, China
| | - Wenjin Wang
- Department of Plastic and Reconstructive Surgery. Shanghai ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Qianru He
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Nantong University, Nantong, JS 226001, China
| | - Huizhu Li
- Sports Medicine Institute of Fudan University, Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China.
| | - Shunyao Li
- Sports Medicine Institute of Fudan University, Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China.
| | - Jian Zhang
- Sports Medicine Institute of Fudan University, Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China.
| | - Yuzhou Chen
- Department of Othopedic Surgery, Xin Hua Hospital affiliated to School of Medicine, Shanghai Jiao Tong University, Shanghai 200092, China
| | - Shuaicheng Guo
- University of Chinese Academy of Sciences, Beijing 100049, China
- Key Laboratory of Infrared System Detection and Imaging Technology, Shanghai Institute of Technical Physics, Chinese Academy of Sciences, Shanghai 200083, China
| | - Lu Fang
- University of Chinese Academy of Sciences, Beijing 100049, China
- Key Laboratory of Infrared System Detection and Imaging Technology, Shanghai Institute of Technical Physics, Chinese Academy of Sciences, Shanghai 200083, China
| | - Stefano Pluchino
- Department of Clinical Neurosciences, University of Cambridge, Cambridge CB2 0AH, UK
| | - Ewelina Biskup
- Department of Basic and Clinical Science, Shanghai University of Medicine and Health Sciences, Shanghai 201318, China
- Department of Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200001, China
| | - Mikhail Artemyev
- Research Institute for Physical Chemical Problems of the Belarusian State University, Leningradskaya srt., 14, Minsk, 220006, Belarus
| | - Fuchun Chen
- Key Laboratory of Infrared System Detection and Imaging Technology, Shanghai Institute of Technical Physics, Chinese Academy of Sciences, Shanghai 200083, China
| | - Yunxia Li
- Sports Medicine Institute of Fudan University, Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China.
| | - Jun Chen
- Sports Medicine Institute of Fudan University, Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China.
| | - Sijia Feng
- Sports Medicine Institute of Fudan University, Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China.
| | - Yan Wo
- Department of Anatomy and Physiology, School of Medicine, Shanghai Jiao Tong University, Shanghai 200025, China.
| |
Collapse
|
29
|
Xu X, Wang Y, Luo X, Gao X, Gu W, Ma Y, Xu L, Yu M, Liu X, Liu J, Wang X, Zheng T, Mao C, Dong L. A non-invasive strategy for suppressing asthmatic airway inflammation and remodeling: Inhalation of nebulized hypoxic hUCMSC-derived extracellular vesicles. Front Immunol 2023; 14:1150971. [PMID: 37090722 PMCID: PMC10113478 DOI: 10.3389/fimmu.2023.1150971] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Accepted: 03/23/2023] [Indexed: 04/08/2023] Open
Abstract
Mesenchymal stromal cell-derived extracellular vesicles (MSC-EVs) are extremely promising nanoscale cell-free therapeutic agents. We previously identified that intravenous administration (IV) of human umbilical cord MSC-EVs (hUCMSC-EVs), especially hypoxic hUCMSC-EVs (Hypo-EVs), could suppress allergic airway inflammation and remodeling. Here, we further investigated the therapeutic effects of Hypo-EVs administration by atomizing inhalation (INH), which is a non-invasive and efficient drug delivery method for lung diseases. We found that nebulized Hypo-EVs produced by the atomization system (medical/household air compressor and nebulizer) maintained excellent structural integrity. Nebulized Dir-labeled Hypo-EVs inhaled by mice were mainly restricted to lungs. INH administration of Hypo-EVs significantly reduced the airway inflammatory infiltration, decreased the levels of IL-4, IL-5 and IL-13 in bronchoalveolar lavage fluid (BALF), declined the content of OVA-specific IgE in serum, attenuated the goblet cell metaplasia, and the expressions of subepithelial collagen-1 and α-smooth muscle actin (α-SMA). Notably, Hypo-EV INH administration was generally more potent than Hypo-EV IV in suppressing IL-13 levels and collagen-1 and α-SMA expressions. RNA sequencing revealed that various biological processes, such as cell adhesion, innate immune response, B cell activation, and extracellular space, were associated with the activity of Hypo-EV INH against asthma mice. In addition, Hypo-EVs could load exogenous miR-146a-5p (miR-146a-5p-EVs). Furthermore, INH administration of miR-146a-5p-EVs resulted in a significantly increased expression of miR-146a-5p mostly in lungs, and offered greater protection against the OVA-induced increase in airway inflammation, subepithelial collagen accumulation and myofibroblast compared with nebulized Hypo-EVs. Overall, nebulized Hypo-EVs effectively attenuated allergic airway inflammation and remodeling, potentially creating a non-invasive route for the use of MSC-EVs in asthma treatment.
Collapse
Affiliation(s)
- Xiaowei Xu
- Department of Nuclear Medicine, The Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Ying Wang
- Department of Respiratory Diseases, The Affiliated Huai’an Hospital of Xuzhou Medical University, Huai’an, Jiangsu, China
| | - Xinkai Luo
- Department of Nuclear Medicine, The Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Xuerong Gao
- Department of Nuclear Medicine, The Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Weifeng Gu
- Department of Nuclear Medicine, The Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Yongbin Ma
- Department of Central Laboratory, Jintan Hospital of Jiangsu University, Changzhou, Jiangsu, China
| | - Lili Xu
- Department of Respiratory Diseases, The Affiliated People’s Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Mengzhu Yu
- Department of Paidology, The Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Xi Liu
- Department of Central Laboratory, The Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Jiameng Liu
- Department of Nuclear Medicine, The Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Xuefeng Wang
- Department of Nuclear Medicine, The Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
- Department of Central Laboratory, The Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Tingting Zheng
- Department of Nuclear Medicine, The Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
- *Correspondence: Liyang Dong, ; Tingting Zheng, ; Chaoming Mao,
| | - Chaoming Mao
- Department of Nuclear Medicine, The Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
- *Correspondence: Liyang Dong, ; Tingting Zheng, ; Chaoming Mao,
| | - Liyang Dong
- Department of Nuclear Medicine, The Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
- *Correspondence: Liyang Dong, ; Tingting Zheng, ; Chaoming Mao,
| |
Collapse
|
30
|
de Assis ACC, Reis ALS, Nunes LV, Ferreira LFR, Bilal M, Iqbal HMN, Soriano RN. Stem Cells and Tissue Engineering-Based Therapeutic Interventions: Promising Strategies to Improve Peripheral Nerve Regeneration. Cell Mol Neurobiol 2023; 43:433-454. [PMID: 35107689 PMCID: PMC11415205 DOI: 10.1007/s10571-022-01199-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Accepted: 01/21/2022] [Indexed: 02/05/2023]
Abstract
Unlike the central nervous system, the peripheral one has the ability to regenerate itself after injury; however, this natural regeneration process is not always successful. In fact, even with some treatments, the prognosis is poor, and patients consequently suffer with the functional loss caused by injured nerves, generating several impacts on their quality of life. In the present review we aimed to address two strategies that may considerably potentiate peripheral nerve regeneration: stem cells and tissue engineering. In vitro studies have shown that pluripotent cells associated with neural scaffolds elaborated by tissue engineering can increase functional recovery, revascularization, remyelination, neurotrophin expression and reduce muscle atrophy. Although these results are very promising, it is important to note that there are some barriers to be circumvented: the host's immune response, the oncogenic properties attributed to stem cells and the duration of the pro-regenerative effects. After all, more studies are still needed to overcome the limitations of these treatments; those that address techniques for manipulating the lesion microenvironment combining different therapies seem to be the most promising and proactive ones.
Collapse
Affiliation(s)
- Ana Carolina Correa de Assis
- Department of Medicine, Federal University of Juiz de Fora (UFJF-GV), 241 Manoel Byrro St., Governador Valadares, MG, 35032-620, Brazil
| | - Amanda Luiza Silva Reis
- Department of Medicine, Federal University of Juiz de Fora (UFJF-GV), 241 Manoel Byrro St., Governador Valadares, MG, 35032-620, Brazil
| | - Leonardo Vieira Nunes
- School of Medicine, Federal University of Juiz de Fora (UFJF-JF), Eugênio do Nascimento Avenue, Juiz de Fora, MG, 36038-330, Brazil
| | - Luiz Fernando Romanholo Ferreira
- Graduate Program in Process Engineering, Tiradentes University (UNIT), 300 Murilo Dantas Ave., Aracaju, SE, 49032-490, Brazil
- Institute of Technology and Research (ITP), Tiradentes University (UNIT), 300 Murilo Dantas Ave., Aracaju, SE, 49032-490, Brazil
| | - Muhammad Bilal
- School of Life Science and Food Engineering, Huaiyin Institute of Technology, Huaian, 223003, China.
| | - Hafiz M N Iqbal
- Tecnologico de Monterrey, School of Engineering and Sciences, Campus Monterrey, Ave. Eugenio Garza Sada 2501, CP 64849, Monterrey, NL , Mexico
| | - Renato Nery Soriano
- Division of Physiology and Biophysics, Department of Basic Life Sciences, Federal University of Juiz de Fora (UFJF-GV), 1167 Moacir Paleta Ave., Governador Valadares, MG, 35020-360, Brazil.
| |
Collapse
|
31
|
Elhessy HM, Habotta OA, Eldesoqui M, Elsaed WM, Soliman MFM, Sewilam HM, Elhassan YH, Lashine NH. Comparative neuroprotective effects of Cerebrolysin, dexamethasone, and ascorbic acid on sciatic nerve injury model: Behavioral and histopathological study. Front Neuroanat 2023; 17:1090738. [PMID: 36816518 PMCID: PMC9928760 DOI: 10.3389/fnana.2023.1090738] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Accepted: 01/05/2023] [Indexed: 02/04/2023] Open
Abstract
Background The majority of the suggested experimental modalities for peripheral nerve injury (PNI) result in varying degrees of recovery in animal models; however, there are not many reliable clinical pharmacological treatment models available. To alleviate PNI complications, research on approaches to accelerate peripheral nerve regeneration is encouraged. Cerebrolysin, dexamethasone, and ascorbic acid (vitamin C) drug models were selected in our study because of their reported curative effects of different mechanisms of action. Methodology A total of 40 adult male albino rats were used in this study. Sciatic nerve crush injury was induced in 32 rats, which were divided equally into four groups (model, Cerebrolysin, dexamethasone, and vitamin C groups) and compared to the sham group (n = 8). The sciatic nerve sensory and motor function regeneration after crushing together with gastrocnemius muscle histopathological changes were evaluated by the sciatic function index, the hot plate test, gastrocnemius muscle mass ratio, and immune expression of S100 and apoptosis cascade (BAX, BCL2, and BAX/BCL2 ratio). Results Significant improvement of the behavioral status and histopathological assessment scores occurred after the use of Cerebrolysin (as a neurotrophic factor), dexamethasone (as an anti-inflammatory), and vitamin C (as an antioxidant). Despite these seemingly concomitant, robust behavioral and pathological changes, vitamin C appeared to have the best results among the three main outcome measures. There was a positive correlation between motor and sensory improvement and also between behavioral and histopathological changes, boosting the effectiveness, and implication of the sciatic function index as a mirror for changes occurring on the tissue level. Conclusion Vitamin C is a promising therapeutic in the treatment of PNI. The sciatic function index (SFI) test is a reliable accurate method for assessing sciatic nerve integrity after both partial disruption and regrowth.
Collapse
Affiliation(s)
- Heba M. Elhessy
- Department of Anatomy and Embryology, Faculty of Medicine, Mansoura University, Mansoura, Egypt,*Correspondence: Heba M. Elhessy,
| | - Ola A. Habotta
- Department of Forensic Medicine and Toxicology, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt
| | - Mamdouh Eldesoqui
- Department of Anatomy and Embryology, Faculty of Medicine, Mansoura University, Mansoura, Egypt,Department of Basic Medical Sciences, College of Medicine, Almaarefa University, Riyadh, Saudi Arabia
| | - Wael M. Elsaed
- Department of Anatomy and Embryology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Mona F. M. Soliman
- Department of Histology and Cell Biology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Haitham M. Sewilam
- Department of Histology and Cell Biology, Faculty of Medicine, Helwan University, Helwan, Egypt
| | - Y. H. Elhassan
- Department of Anatomy, College of Medicine, Taibah University, Medina, Saudi Arabia
| | - Nermeen H. Lashine
- Department of Anatomy and Embryology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| |
Collapse
|
32
|
Sanz-Ros J, Mas-Bargues C, Romero-García N, Huete-Acevedo J, Dromant M, Borrás C. Extracellular Vesicles as Therapeutic Resources in the Clinical Environment. Int J Mol Sci 2023; 24:2344. [PMID: 36768664 PMCID: PMC9917082 DOI: 10.3390/ijms24032344] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 01/19/2023] [Accepted: 01/21/2023] [Indexed: 01/26/2023] Open
Abstract
The native role of extracellular vesicles (EVs) in mediating the transfer of biomolecules between cells has raised the possibility to use them as therapeutic vehicles. The development of therapies based on EVs is now expanding rapidly; here we will describe the current knowledge on different key points regarding the use of EVs in a clinical setting. These points are related to cell sources of EVs, isolation, storage, and delivery methods, as well as modifications to the releasing cells for improved production of EVs. Finally, we will depict the application of EVs therapies in clinical trials, considering the impact of the COVID-19 pandemic on the development of these therapies, pointing out that although it is a promising therapy for human diseases, we are still in the initial phase of its application to patients.
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
- Department of Cardiology, 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
- Department of Anesthesiology and Surgical Trauma Intensive Care, Hospital Clinic Universitari de Valencia, University of Valencia, 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
| |
Collapse
|
33
|
Pan C, Xu P, Zheng Y, Wang Y, Chen C, Fu S, Liu Z, Chen Y, Xue K, Zhou Q, Liu K. Preparation of therapy-grade extracellular vesicles from adipose tissue to promote diabetic wound healing. Front Bioeng Biotechnol 2023; 11:1129187. [PMID: 37034267 PMCID: PMC10076785 DOI: 10.3389/fbioe.2023.1129187] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 03/10/2023] [Indexed: 04/11/2023] Open
Abstract
Background: Treatment of diabetic wounds is a major challenge in clinical practice. Extracellular vesicles (EVs) from adipose-derived stem cells have shown effectiveness in diabetic wound models. However, obtaining ADSC-EVs requires culturing vast numbers of cells, which is hampered by the need for expensive equipment and reagents, extended time cost, and complicated procedures before commercialization. Therefore, methods to extract EVs from discarded tissue need to be developed, for immediate application during surgery. For this reason, mechanical, collagenase-digestive, and constant in-vitro-collective methods were designed and compared for preparing therapy-grade EVs directly from adipose tissue. Methods: Characteristics and quantities of EVs were detected by transmission electron microscopy, nanoparticle tracking analysis, and Western blotting firstly. To investigate the biological effects of EVs on diabetic wound healing, angiogenesis, proliferation, migration, and inflammation-regulation assays were then evaluated in vitro, along with a diabetic wound healing mouse model in vivo. To further explore the potential therapeutic mechanism of EVs, miRNA expression profile of EVs were also identified and analyzed. Results: The adipose tissue derived EVs (AT-EVs) were showed to qualify ISEV identification by nanoparticle tracking analysis and Western blotting and the AT-EVs yield from three methods was equal. EVs also showed promoting effects on biological processes related to diabetic wound healing, which depend on fibroblasts, keratinocytes, endothelial cells, and macrophages both in vitro and in vivo. We also observed enrichment of overlapping or unique miRNAs originate from different types of AT-EVs associated with diabetic wound healing for further investigation. Conclusion: After comparative analyses, a mechanical method was proposed for preparing immediate clinical applicable EVs from adipose tissue that would result in reduced preparation time and lower cost, which could have promising application potential in treating diabetic wounds.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Kai Liu
- *Correspondence: Qimin Zhou, ; Kai Liu,
| |
Collapse
|
34
|
Reshamwala R, Shah M. Regenerative Approaches in the Nervous System. Regen Med 2023. [DOI: 10.1007/978-981-19-6008-6_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
|
35
|
Han N, Zhang W, Fang XX, Li QC, Pi W. Reduced graphene oxide-embedded nerve conduits loaded with bone marrow mesenchymal stem cell-derived extracellular vesicles promote peripheral nerve regeneration. Neural Regen Res 2023. [PMID: 35799543 PMCID: PMC9241414 DOI: 10.4103/1673-5374.343889] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
We previously combined reduced graphene oxide (rGO) with gelatin-methacryloyl (GelMA) and polycaprolactone (PCL) to create an rGO-GelMA-PCL nerve conduit and found that the conductivity and biocompatibility were improved. However, the rGO-GelMA-PCL nerve conduits differed greatly from autologous nerve transplants in their ability to promote the regeneration of injured peripheral nerves and axonal sprouting. Extracellular vesicles derived from bone marrow mesenchymal stem cells (BMSCs) can be loaded into rGO-GelMA-PCL nerve conduits for repair of rat sciatic nerve injury because they can promote angiogenesis at the injured site. In this study, 12 weeks after surgery, sciatic nerve function was measured by electrophysiology and sciatic nerve function index, and myelin sheath and axon regeneration were observed by electron microscopy, immunohistochemistry, and immunofluorescence. The regeneration of microvessel was observed by immunofluorescence. Our results showed that rGO-GelMA-PCL nerve conduits loaded with BMSC-derived extracellular vesicles were superior to rGO-GelMA-PCL conduits alone in their ability to increase the number of newly formed vessels and axonal sprouts at the injury site as well as the recovery of neurological function. These findings indicate that rGO-GelMA-PCL nerve conduits loaded with BMSC-derived extracellular vesicles can promote peripheral nerve regeneration and neurological function recovery, and provide a new direction for the curation of peripheral nerve defect in the clinic.
Collapse
|
36
|
Ng WC, Lokanathan Y, Baki MM, Fauzi MB, Zainuddin AA, Azman M. Tissue Engineering as a Promising Treatment for Glottic Insufficiency: A Review on Biomolecules and Cell-Laden Hydrogel. Biomedicines 2022; 10:3082. [PMID: 36551838 PMCID: PMC9775346 DOI: 10.3390/biomedicines10123082] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 11/11/2022] [Accepted: 11/16/2022] [Indexed: 12/03/2022] Open
Abstract
Glottic insufficiency is widespread in the elderly population and occurs as a result of secondary damage or systemic disease. Tissue engineering is a viable treatment for glottic insufficiency since it aims to restore damaged nerve tissue and revitalize aging muscle. After injection into the biological system, injectable biomaterial delivers cost- and time-effectiveness while acting as a protective shield for cells and biomolecules. This article focuses on injectable biomaterials that transport cells and biomolecules in regenerated tissue, particularly adipose, muscle, and nerve tissue. We propose Wharton's Jelly mesenchymal stem cells (WJMSCs), induced pluripotent stem cells (IP-SCs), basic fibroblast growth factor (bFGF), vascular endothelial growth factor (VEGF), hepatocyte growth factor (HGF), insulin growth factor-1 (IGF-1) and extracellular vesicle (EV) as potential cells and macromolecules to be included into biomaterials, with some particular testing to support them as a promising translational medicine for vocal fold regeneration.
Collapse
Affiliation(s)
- Wan-Chiew Ng
- Department of Otorhinolaryngology-Head and Neck Surgery, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
| | - Yogeswaran Lokanathan
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
| | - Marina Mat Baki
- Department of Otorhinolaryngology-Head and Neck Surgery, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
| | - Mh Busra Fauzi
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
| | - Ani Amelia Zainuddin
- Department of Obstetrics and Gynaecology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
| | - Mawaddah Azman
- Department of Otorhinolaryngology-Head and Neck Surgery, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
| |
Collapse
|
37
|
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
|
38
|
Xue T, Wang X, Ru J, Zhang L, Yin H. The inhibitory effect of human umbilical cord mesenchymal stem cells expressing anti-HAAH scFv-sTRAIL fusion protein on glioma. Front Bioeng Biotechnol 2022; 10:997799. [DOI: 10.3389/fbioe.2022.997799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 10/26/2022] [Indexed: 11/10/2022] Open
Abstract
Glioma is the most common malignant intracranial tumor with low 5-year survival rate. In this study, we constructed a plasmid expressing anti-HAAH single-chain antibody and sTRAIL fusion protein (scFv-sTRAIL), and explored the effects of the double gene modified human umbilical cord mesenchyreal stem cells (hucMSCs) on the growth of glioma in vitro and in vivo. The isolated hucMSCs were identified by detecting the adipogenic differentiation ability and the osteogenic differentiation ability. The phenotypes of hucMSCs were determined by the flow cytometry. The hucMSCs were infected with lentivirus expression scFv-sTRAIL fusion protein. The expression of sTRAIL in hucMSCs were detected by immunofluorescence staining, western blot and ELISA. The tropism of hucMSCs toward U87G cells was assessed by transwell assay. The inhibitory effect of hucMSCs on U87G cells were explored by CCK8 and apoptosis assay. The xenograft tumor was established by subcutaneously injection of U87G cells into the back of mice. The hucMSCs were injected via tail veins. The inhibitory effect of hucMSCs on glioma in vivo was assessed by TUNEL assay. The hucMSCs migrated into the xenograft tumor were revealed by detecting the green fluorescent. The results showed that the scFv-sTRAIL expression did not affect the phenotypes of hucMSCs. The scFv-sTRAIL expression promoted the tropism of hucMSCs toward U87G cells, enhanced the inhibitory effect and tumor killing effect of hucMSCs on U87G cells. The in vivo study showed that hucMSCs expressing scFv-sTRAIL demonstrated significantly higher inhibitory effect and tumor killing effect than hucMSCs expressing sTRAIL. The green fluorescence intensity in the mice injected with hucMSCs expressing scFv-sTRAIL was significantly higher than that injected with hucMSCs expressing sTRAIL. These data suggested that the scFv conferred the targeting effect of hucMSCs tropism towards the xenograft tumor. In conclusion, the hucMSCs expressing scFv-sTRAIL fusion protein gained the capability to target and kill gliomas cells in vitro and in vivo. These findings shed light on a potential therapy for glioma treatment.
Collapse
|
39
|
Gál P, Brábek J, Holub M, Jakubek M, Šedo A, Lacina L, Strnadová K, Dubový P, Hornychová H, Ryška A, Smetana K. Autoimmunity, cancer and COVID-19 abnormally activate wound healing pathways: critical role of inflammation. Histochem Cell Biol 2022; 158:415-434. [PMID: 35867145 PMCID: PMC9305064 DOI: 10.1007/s00418-022-02140-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/06/2022] [Indexed: 11/30/2022]
Abstract
Recent evidence indicates that targeting IL-6 provides broad therapeutic approaches to several diseases. In patients with cancer, autoimmune diseases, severe respiratory infections [e.g. coronavirus disease 2019 (COVID-19)] and wound healing, IL-6 plays a critical role in modulating the systemic and local microenvironment. Elevated serum levels of IL-6 interfere with the systemic immune response and are associated with disease progression and prognosis. As already noted, monoclonal antibodies blocking either IL-6 or binding of IL-6 to receptors have been used/tested successfully in the treatment of rheumatoid arthritis, many cancer types, and COVID-19. Therefore, in the present review, we compare the impact of IL-6 and anti-IL-6 therapy to demonstrate common (pathological) features of the studied diseases such as formation of granulation tissue with the presence of myofibroblasts and deposition of new extracellular matrix. We also discuss abnormal activation of other wound-healing-related pathways that have been implicated in autoimmune disorders, cancer or COVID-19.
Collapse
Affiliation(s)
- Peter Gál
- Department of Pharmacology, Pavol Jozef Šafárik University, Košice, Slovak Republic
- Department of Biomedical Research, East-Slovak Institute of Cardiovascular Diseases, Košice, Slovak Republic
- Prague Burn Centre, Third Faculty of Medicine, Charles University and University Hospital Kralovske Vinohrady, Prague, Czech Republic
| | - Jan Brábek
- Department of Cell Biology, Faculty of Science, Charles University, 120 00 Prague 2, Czech Republic
- BIOCEV, Faculty of Science, Charles University, 252 50 Vestec, Czech Republic
| | - Michal Holub
- Department of Infectious Diseases, First Faculty of Medicine, Military University Hospital Prague and Charles University, 160 00 Prague, Czech Republic
| | - Milan Jakubek
- Department of Paediatrics and Adolescent Medicine, First Faculty of Medicine, Charles University, 120 00 Prague 2, Czech Republic
- BIOCEV, First Faculty of Medicine, Charles University, 252 50 Vestec, Czech Republic
- Department of Analytical Chemistry, University of Chemistry and Technology Prague, 166 28 Prague 6, Czech Republic
| | - Aleksi Šedo
- Institute of Biochemistry and Experimental Oncology, First Faculty of Medicine, Charles University, 120 00 Praha 2, Czech Republic
| | - Lukáš Lacina
- BIOCEV, First Faculty of Medicine, Charles University, 252 50 Vestec, Czech Republic
- Institute of Anatomy, First Faculty of Medicine, Charles University, 120 00 Prague 2, Czech Republic
- Department of Dermatovenereology, First Faculty of Medicine, Charles University, 120 00 Prague 2, Czech Republic
| | - Karolína Strnadová
- BIOCEV, First Faculty of Medicine, Charles University, 252 50 Vestec, Czech Republic
- Institute of Anatomy, First Faculty of Medicine, Charles University, 120 00 Prague 2, Czech Republic
| | - Petr Dubový
- Institute of Anatomy, Faculty of Medicine, Masaryk University, 625 00 Brno, Czech Republic
| | - Helena Hornychová
- The Fingerland Department of Pathology, Faculty of Medicine Hradec Králové, Charles University, 500 05 Hradec Králové, Czech Republic
| | - Aleš Ryška
- The Fingerland Department of Pathology, Faculty of Medicine Hradec Králové, Charles University, 500 05 Hradec Králové, Czech Republic
| | - Karel Smetana
- BIOCEV, First Faculty of Medicine, Charles University, 252 50 Vestec, Czech Republic
- Institute of Anatomy, First Faculty of Medicine, Charles University, 120 00 Prague 2, Czech Republic
| |
Collapse
|
40
|
McLaughlin C, Datta P, Singh YP, Lo A, Horchler S, Elcheva IA, Ozbolat IT, Ravnic DJ, Koduru SV. Mesenchymal Stem Cell-Derived Extracellular Vesicles for Therapeutic Use and in Bioengineering Applications. Cells 2022; 11:3366. [PMID: 36359762 PMCID: PMC9657427 DOI: 10.3390/cells11213366] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 10/10/2022] [Accepted: 10/19/2022] [Indexed: 07/25/2023] Open
Abstract
Extracellular vesicles (EVs) are small lipid bilayer-delimited particles that are naturally released from cells into body fluids, and therefore can travel and convey regulatory functions in the distal parts of the body. EVs can transmit paracrine signaling by carrying over cytokines, chemokines, growth factors, interleukins (ILs), transcription factors, and nucleic acids such as DNA, mRNAs, microRNAs, piRNAs, lncRNAs, sn/snoRNAs, mtRNAs and circRNAs; these EVs travel to predecided destinations to perform their functions. While mesenchymal stem cells (MSCs) have been shown to improve healing and facilitate treatments of various diseases, the allogenic use of these cells is often accompanied by serious adverse effects after transplantation. MSC-produced EVs are less immunogenic and can serve as an alternative to cellular therapies by transmitting signaling or delivering biomaterials to diseased areas of the body. This review article is focused on understanding the properties of EVs derived from different types of MSCs and MSC-EV-based therapeutic options. The potential of modern technologies such as 3D bioprinting to advance EV-based therapies is also discussed.
Collapse
Affiliation(s)
- Caroline McLaughlin
- Department of Surgery, Penn State Health Milton S. Hershey Medical Center, Hershey, PA 17033, USA
| | - Pallab Datta
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER) Kolkata, West Bengal 700054, India
| | - Yogendra P. Singh
- Department of Biomedical Engineering, Materials Research Institute, The Huck Institutes of Life Sciences, Penn State University, University Park, PA 16802, USA
| | - Alexis Lo
- Department of Surgery, Penn State Health Milton S. Hershey Medical Center, Hershey, PA 17033, USA
| | - Summer Horchler
- Department of Surgery, Penn State Health Milton S. Hershey Medical Center, Hershey, PA 17033, USA
| | - Irina A. Elcheva
- Department of Pediatrics, Hematology/Oncology, Penn State College of Medicine, Hershey, PA 17033, USA
| | - Ibrahim T. Ozbolat
- Department of Biomedical Engineering, Materials Research Institute, The Huck Institutes of Life Sciences, Penn State University, University Park, PA 16802, USA
| | - Dino J. Ravnic
- Department of Surgery, Penn State Health Milton S. Hershey Medical Center, Hershey, PA 17033, USA
| | - Srinivas V. Koduru
- Department of Surgery, Penn State Health Milton S. Hershey Medical Center, Hershey, PA 17033, USA
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, PA 17033, USA
| |
Collapse
|
41
|
Pozzobon M, D’Agostino S, Roubelakis MG, Cargnoni A, Gramignoli R, Wolbank S, Gindraux F, Bollini S, Kerdjoudj H, Fenelon M, Di Pietro R, Basile M, Borutinskaitė V, Piva R, Schoeberlein A, Eissner G, Giebel B, Ponsaerts P. General consensus on multimodal functions and validation analysis of perinatal derivatives for regenerative medicine applications. Front Bioeng Biotechnol 2022; 10:961987. [PMID: 36263355 PMCID: PMC9574482 DOI: 10.3389/fbioe.2022.961987] [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: 06/05/2022] [Accepted: 07/01/2022] [Indexed: 11/26/2022] Open
Abstract
Perinatal tissues, such as placenta and umbilical cord contain a variety of somatic stem cell types, spanning from the largely used hematopoietic stem and progenitor cells to the most recently described broadly multipotent epithelial and stromal cells. As perinatal derivatives (PnD), several of these cell types and related products provide an interesting regenerative potential for a variety of diseases. Within COST SPRINT Action, we continue our review series, revising and summarizing the modalities of action and proposed medical approaches using PnD products: cells, secretome, extracellular vesicles, and decellularized tissues. Focusing on the brain, bone, skeletal muscle, heart, intestinal, liver, and lung pathologies, we discuss the importance of potency testing in validating PnD therapeutics, and critically evaluate the concept of PnD application in the field of tissue regeneration. Hereby we aim to shed light on the actual therapeutic properties of PnD, with an open eye for future clinical application. This review is part of a quadrinomial series on functional/potency assays for validation of PnD, spanning biological functions, such as immunomodulation, anti-microbial/anti-cancer, anti-inflammation, wound healing, angiogenesis, and regeneration.
Collapse
Affiliation(s)
- Michela Pozzobon
- Department of Women’s and Children’s Health, University of Padova, Padova, Italy
| | - Stefania D’Agostino
- Department of Women’s and Children’s Health, University of Padova, Padova, Italy
| | - Maria G. Roubelakis
- Laboratory of Biology, Medical School of Athens, National and Kapodistrian University of Athens, Athens, Greece
| | - Anna Cargnoni
- Centro di Ricerca E. Menni, Fondazione Poliambulanza Istituto Ospedaliero, Brescia, Italy
| | - Roberto Gramignoli
- Department of Laboratory Medicine, Division of Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Susanne Wolbank
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, The Research Center in Cooperation with AUVA Trauma Research Center, Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Florelle Gindraux
- Service de Chirurgie Orthopédique, Traumatologique et plastique, CHU Besançon, Laboratoire de Nanomédecine, Imagerie, Thérapeutique EA 4662, University Bourgogne Franche-Comté, Besançon, France
| | - Sveva Bollini
- Department of Experimental Medicine (DIMES), School of Medical and Pharmaceutical Sciences, University of Genova, Genova, Italy
| | - Halima Kerdjoudj
- University of Reims Champagne Ardenne, EA 4691 BIOS “Biomatériaux et Inflammation en Site Osseux”, UFR d’Odontologie, Reims, France
| | | | - Roberta Di Pietro
- Department of Medicine and Ageing Sciences, Section of Biomorphology, G. d'Annunzio University of Chieti-Pescara, Chieti, Italy
| | - Mariangela Basile
- Department of Medicine and Ageing Sciences, Section of Biomorphology, G. d'Annunzio University of Chieti-Pescara, Chieti, Italy
| | - Veronika Borutinskaitė
- Department of Molecular Cell Biology, Institute of Biochemistry, Vilnius University, Vilnius, Lithuania
| | - Roberta Piva
- Department of Neuroscience and Rehabilitation, University of Ferrara, Ferrara, Italy
| | - Andreina Schoeberlein
- Department of Obstetrics and Feto-maternal Medicine, Inselspital, Bern University Hospital, Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Guenther Eissner
- Systems Biology Ireland, School of Medicine, Conway Institute, University College Dublin, Dublin, Ireland
| | - Bernd Giebel
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Peter Ponsaerts
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Antwerp, Belgium
| |
Collapse
|
42
|
Liu B, Kong Y, Shi W, Kuss M, Liao K, Hu G, Xiao P, Sankarasubramanian J, Guda C, Wang X, Lei Y, Duan B. Exosomes derived from differentiated human ADMSC with the Schwann cell phenotype modulate peripheral nerve-related cellular functions. Bioact Mater 2022; 14:61-75. [PMID: 35310346 PMCID: PMC8892082 DOI: 10.1016/j.bioactmat.2021.11.022] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Revised: 11/09/2021] [Accepted: 11/23/2021] [Indexed: 02/07/2023] Open
Abstract
Peripheral nerve regeneration remains a significant clinical challenge due to the unsatisfactory functional recovery and public health burden. Exosomes, especially those derived from mesenchymal stem cells (MSCs), are promising as potential cell-free therapeutics and gene therapy vehicles for promoting neural regeneration. In this study, we reported the differentiation of human adipose derived MSCs (hADMSCs) towards the Schwann cell (SC) phenotype (hADMSC-SCs) and then isolated exosomes from hADMSCs with and without differentiation (i.e., dExo vs uExo). We assessed and compared the effects of uExo and dExo on antioxidative, angiogenic, anti-inflammatory, and axon growth promoting properties by using various peripheral nerve-related cells. Our results demonstrated that hADMSC-SCs secreted more neurotrophic factors and other growth factors, compared to hADMSCs without differentiation. The dExo isolated from hADMSC-SCs protected rat SCs from oxidative stress and enhanced HUVEC migration and angiogenesis. Compared to uExo, dExo also had improved performances in downregulating pro-inflammatory gene expressions and cytokine secretions and promoting axonal growth of sensory neurons differentiated from human induced pluripotent stem cells. Furthermore, microRNA (miRNA) sequencing analysis revealed that exosomes and their parent cells shared some similarities in their miRNA profiles and exosomes displayed a distinct miRNA signature. Many more miRNAs were identified in dExo than in uExo. Several upregulated miRNAs, like miRNA-132-3p and miRNA-199b-5p, were highly related to neuroprotection, anti-inflammation, and angiogenesis. The dExo can effectively modulate various peripheral nerve-related cellular functions and is promising for cell-free biological therapeutics to enhance neural regeneration. Exosomes were isolated from hADMSCs with and without differentiation towards SC phenotype (i.e., dExo vs uExo). hADMSC-SCs secreted more growth factors compared to hADMSCs without differentiation. The dExo protected rat SCs from oxidative stress and enhanced endothelial cell migration and angiogenesis. dExo promoted axonal growth of sensory neurons differentiated from hiPSCs. miRNA sequencing analysis unveiled and compared the exosomal and cellular miRNA profiles.
Collapse
|
43
|
Huang Y, Zhang Y, He Z, Manyande A, Wu D, Feng M, Xiang H. The connectome from the cerebral cortex to skeletal muscle using viral transneuronal tracers: a review. Am J Transl Res 2022; 14:4864-4879. [PMID: 35958450 PMCID: PMC9360884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Accepted: 06/22/2022] [Indexed: 06/15/2023]
Abstract
Connectomics has developed from an initial observation under an electron microscope to the present well-known medical imaging research approach. The emergence of the most popular transneuronal tracers has further advanced connectomics research. Researchers use the virus trans-nerve tracing method to trace the whole brain, mark the brain nerve circuit and nerve connection structure, and construct a complete nerve conduction pathway. This review assesses current methods of studying cortical to muscle connections using viral neuronal tracers and demonstrates their application in disease diagnosis and prognosis.
Collapse
Affiliation(s)
- Yan Huang
- Tongji Hospital of Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430030, Hubei, P. R. China
- Department of Interventional Therapy, The First Affiliated Hospital of Dalian Medical UniversityDalian 116000, Liaoning, P. R. China
| | - Yunhua Zhang
- Hubei Provincial Hospital of Traditional Chinese MedicineWuhan 430061, Hubei, P. R. China
- Clinical Medical College of Hubei University of Chinese MedicineWuhan 430061, Hubei, P. R. China
- Hubei Province Academy of Traditional Chinese MedicineWuhan 430061, Hubei, P. R. China
| | - Zhigang He
- Tongji Hospital of Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430030, Hubei, P. R. China
| | - Anne Manyande
- School of Human and Social Sciences, University of West LondonLondon, UK
| | - Duozhi Wu
- Department of Anesthesiology, Hainan General HospitalHaikou 570311, Hainan, P. R. China
| | - Maohui Feng
- Department of Gastrointestinal Surgery, Wuhan Peritoneal Cancer Clinical Medical Research Center, Zhongnan Hospital of Wuhan University, Hubei Key Laboratory of Tumor Biological Behaviors and Hubei Cancer Clinical Study CenterWuhan 430071, Hubei, P. R. China
| | - Hongbing Xiang
- Tongji Hospital of Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430030, Hubei, P. R. China
| |
Collapse
|
44
|
Bahmani L, Ullah M. Different Sourced Extracellular Vesicles and Their Potential Applications in Clinical Treatments. Cells 2022; 11:1989. [PMID: 35805074 PMCID: PMC9265969 DOI: 10.3390/cells11131989] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Revised: 06/19/2022] [Accepted: 06/20/2022] [Indexed: 12/12/2022] Open
Abstract
Extracellular vesicles (EVs) include a heterogeneous group of natural cell-derived nanostructures that are increasingly regarded as promising biotherapeutic agents and drug delivery vehicles in human medicine. Desirable intrinsic properties of EVs including the ability to bypass natural membranous barriers and to deliver their unique biomolecular cargo to specific cell populations position them as fiercely competitive alternatives for currently available cell therapies and artificial drug delivery platforms. EVs with distinct characteristics can be released from various cell types into the extracellular environment as a means of transmitting bioactive components and altering the status of the target cell. Despite the existence of a large number of preclinical studies confirming the therapeutic efficacy of different originated EVs for treating several pathological conditions, in this review, we first provide a brief overview of EV biophysical properties with an emphasis on their intrinsic therapeutic benefits over cell-based therapies and synthetic delivery systems. Next, we describe in detail different EVs derived from distinct cell sources, compare their advantages and disadvantages, and recapitulate their therapeutic effects on various human disorders to highlight the progress made in harnessing EVs for clinical applications. Finally, knowledge gaps and concrete hurdles that currently hinder the clinical translation of EV therapies are debated with a futuristic perspective.
Collapse
Affiliation(s)
- Leila Bahmani
- Institute for Immunity and Transplantation, Stem Cell Biology and Regenerative Medicine, School of Medicine, Stanford University, Palo Alto, CA 94304, USA;
- Molecular Medicine Department of Medicine, Stanford University, Palo Alto, CA 94304, USA
| | - Mujib Ullah
- Institute for Immunity and Transplantation, Stem Cell Biology and Regenerative Medicine, School of Medicine, Stanford University, Palo Alto, CA 94304, USA;
- Molecular Medicine Department of Medicine, Stanford University, Palo Alto, CA 94304, USA
| |
Collapse
|
45
|
Liu X, Hu L, Liu F. Mesenchymal stem cell-derived extracellular vesicles for cell-free therapy of ocular diseases. EXTRACELLULAR VESICLES AND CIRCULATING NUCLEIC ACIDS 2022; 3:102-117. [PMID: 39698446 PMCID: PMC11648472 DOI: 10.20517/evcna.2022.08] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 03/31/2022] [Accepted: 04/18/2022] [Indexed: 12/20/2024]
Abstract
Mesenchymal stem cells-derived extracellular vesicles (MSC-EVs) have noticeably attracted clinicians' attention in treating ocular diseases. As the paracrine factor of MSCs and an alternative for cell-free therapies, MSC-EVs can be conveniently dropped over the ocular surface or diffused through the retina upon intravitreal injection, without increasing the risks of cellular rejection and tumor formation. For clinical translation, a standardized and scalable production, as well as reprogramming the MSC-EVs, are highly encouraged. This review aims to assess the potential approaches for EV production and functional modification, in addition to summarizing the worldwide clinical trials initiated for various physiological systems and the specific biochemical effects of MSC-EVs on the therapy of eye diseases. Recent advances in the therapy of ocular diseases based on MSC-EVs are reviewed, and the associated challenges and prospects are discussed as well.
Collapse
Affiliation(s)
- Xiaoling Liu
- Eye Hospital, School of Ophthalmology and Optometry, School of Biomedical Engineering, Wenzhou Medical University, Wenzhou 325000, Zhejiang, China
| | - Liang Hu
- Eye Hospital, School of Ophthalmology and Optometry, School of Biomedical Engineering, Wenzhou Medical University, Wenzhou 325000, Zhejiang, China
| | - Fei Liu
- Eye Hospital, School of Ophthalmology and Optometry, School of Biomedical Engineering, Wenzhou Medical University, Wenzhou 325000, Zhejiang, China
- Wenzhou Institute, University of Chinese Academy of Science, Wenzhou 325000, Zhejiang, China
| |
Collapse
|
46
|
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
|
47
|
Yan C, Yu J. Noncoding RNA in Extracellular Vesicles Regulate Differentiation of Mesenchymal Stem Cells. FRONTIERS IN DENTAL MEDICINE 2022. [DOI: 10.3389/fdmed.2021.806001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
To achieve the desired outcome in tissue engineering regeneration, mesenchymal stem cells need to undergo a series of biological processes, including differentiating into the ideal target cells. The extracellular vesicle (EV) in the microenvironment contributes toward determining the fate of the cells with epigenetic regulation, particularly from noncoding RNA (ncRNA), and exerts transportation and protective effects on ncRNAs. We focused on the components and functions of ncRNA (particularly microRNA) in the EVs. The EVs modified by the ncRNA favor tissue regeneration and pose a potential challenge.
Collapse
|
48
|
Thomas SC, Kim JW, Pauletti GM, Hassett DJ, Kotagiri N. Exosomes: Biological Pharmaceutical Nanovectors for Theranostics. Front Bioeng Biotechnol 2022; 9:808614. [PMID: 35096795 PMCID: PMC8790084 DOI: 10.3389/fbioe.2021.808614] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 12/24/2021] [Indexed: 12/12/2022] Open
Abstract
Exosomes are natural cell-derived nanovesicles of endocytic origin that enable cellular crosstalk by transferring encapsulated molecular cargos across biological barriers, thereby holding significantly complex implications in the etiology and progression of diverse disease states. Consequently, the development of exosomes-based nano-theranostic strategies has received immense consideration for advancing therapeutic interventions and disease prognosis. Their favorable biopharmaceutical properties make exosomes a unique nanoparticulate carrier for pharmaceutical drug delivery. This review provides an update on the contemporary strategies utilizing exosomes for theranostic applications in nanomedicine. In addition, we provide a synopsis of exosomal features and insights into strategic modifications that control in vivo biodistribution. We further discuss their opportunities, merits and pitfalls for cell/tissue targeted drug delivery in personalized nanotherapy.
Collapse
Affiliation(s)
- Shindu C Thomas
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, OH, United States
| | - Jin-Woo Kim
- Department of Biological and Agricultural Engineering, Institute for Nanoscience and Engineering, University of Arkansas, Fayetteville, AR, United States
| | - Giovanni M Pauletti
- St. Louis College of Pharmacy, University of Health Sciences and Pharmacy in St. Louis, St. Louis, MO, United States
| | - Daniel J Hassett
- Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Nalinikanth Kotagiri
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, OH, United States
| |
Collapse
|
49
|
Qian W, Li G. Extracellular Vesicles from Esophageal Squamous Cell Carcinoma Promote Angiogenesis and Tumor Growth. J BIOMATER TISS ENG 2022. [DOI: 10.1166/jbt.2022.2896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Angiogenesis is a prerequisite for tumor development and metastasis. Emerging evidence shows that tumor-derived extracellular vesicles (EVs) are an important component of tumor microenvironment, which participate in the communication between normal cells and tumor cells. In this study,
we aimed to investigate the role of EVs derived from esophageal squamous cell carcinoma (ESCC) on tumor angiogenesis. We found that ESCC cell-derived EVs promoted the proliferation, migration, and tubule formation of human umbilical vein endothelial cells in vitro, and enhanced angiogenesis
and tumor growth in vivo. Our results suggest that ESCC cell-derived EVs could promote angio-genesis and tumor growth, which also indicated the application of EVs as a valuable therapeutic strategy of ESCC.
Collapse
Affiliation(s)
- Wenxiu Qian
- Department of Pediatrics, Jintan Hospital, Jiangsu University, Jintan, Changzhou, Jiangsu 213200, P. R. China
| | - Guomin Li
- Department of Critical Care Medicine, Jintan Hospital, Jiangsu University, Jintan, Changzhou, Jiangsu 213200, P. R. China
| |
Collapse
|
50
|
Li X, Guan Y, Li C, Zhang T, Meng F, Zhang J, Li J, Chen S, Wang Q, Wang Y, Peng J, Tang J. Immunomodulatory effects of mesenchymal stem cells in peripheral nerve injury. Stem Cell Res Ther 2022; 13:18. [PMID: 35033187 PMCID: PMC8760713 DOI: 10.1186/s13287-021-02690-2] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 12/18/2021] [Indexed: 12/14/2022] Open
Abstract
Various immune cells and cytokines are present in the aftermath of peripheral nerve injuries (PNI), and coordination of the local inflammatory response is of great significance for the recovery of PNI. Mesenchymal stem cells (MSCs) exhibit immunosuppressive and anti-inflammatory abilities which can accelerate tissue regeneration and attenuate inflammation, but the role of MSCs in the regulation of the local inflammatory microenvironment after PNI has not been widely studied. Here, we summarize the known interactions between MSCs, immune cells, and inflammatory cytokines following PNI with a focus on the immunosuppressive role of MSCs. We also discuss the immunomodulatory potential of MSC-derived extracellular vesicles as a new cell-free treatment for PNI.
Collapse
Affiliation(s)
- Xiangling Li
- The Fourth Medical Center of Chinese PLA General Hospital, Beijing, 100853, People's Republic of China.,Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, 100853, People's Republic of China.,The School of Medicine, Jinzhou Medical University, Jinzhou, 121099, People's Republic of China
| | - Yanjun Guan
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, 100853, People's Republic of China
| | - Chaochao Li
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, 100853, People's Republic of China
| | - Tieyuan Zhang
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, 100853, People's Republic of China
| | - Fanqi Meng
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, 100853, People's Republic of China.,Department of Spine Surgery, Peking University People's Hospital, Beijing, 100044, People's Republic of China
| | - Jian Zhang
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, 100853, People's Republic of China
| | - Junyang Li
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, 100853, People's Republic of China.,The School of Medicine, Nankai University, Tianjin, 300071, People's Republic of China
| | - Shengfeng Chen
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, 100853, People's Republic of China
| | - Qi Wang
- The Fourth Medical Center of Chinese PLA General Hospital, Beijing, 100853, People's Republic of China.,The School of Medicine, Jinzhou Medical University, Jinzhou, 121099, People's Republic of China
| | - Yi Wang
- Department of Stomatology, First Medical Center, Chinese PLA General Hospital, Beijing, 100853, People's Republic of China.
| | - Jiang Peng
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, 100853, People's Republic of China.
| | - Jinshu Tang
- The Fourth Medical Center of Chinese PLA General Hospital, Beijing, 100853, People's Republic of China.
| |
Collapse
|