51
|
Human Adipose Mesenchymal Stem Cell-Derived Exosomes: A Key Player in Wound Healing. Tissue Eng Regen Med 2021; 18:537-548. [PMID: 33547566 DOI: 10.1007/s13770-020-00316-x] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 10/05/2020] [Accepted: 10/29/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Human adipose-derived mesenchymal stem cells (AMSCs) are an attractive resource for wound healing because their regenerative capacity improves injury repair. Recently, stem cell-derived exosomes have been shown to play a positive role in stem cell-based therapies. However, the effects of exosomes derived from AMSCs (AEXOs) on wound healing are unclear. In this study, we aimed to examine the role of AEXOs in attenuating inflammation and explore their effects in normal wound healing. METHODS We isolated exosomes from AMSCs and established a cellular model of inflammation by treatment with the inflammatory cytokines, interferon gamma and tumor necrosis factor alpha, to determine whether AEXOs can inhibit inflammation. We examined the wound healing effects of AEXOs in in vitro wound healing models and performed a miRNA array to understand the role of AEXOs in inflammation and wound healing. RESULTS A significant difference was observed in wound closure and the expression of anti-inflammatory and wound-healing-related factors between control and AEXO-treated cells. CONCLUSION Our results showed that besides alleviating the inflammation response, AEXOs also promote wound healing. Thus, AEXOs represent a novel, stem-cell-based, therapeutic strategy for wound healing.
Collapse
|
52
|
Zhang L, Ouyang P, He G, Wang X, Song D, Yang Y, He X. Exosomes from microRNA-126 overexpressing mesenchymal stem cells promote angiogenesis by targeting the PIK3R2-mediated PI3K/Akt signalling pathway. J Cell Mol Med 2021; 25:2148-2162. [PMID: 33350092 PMCID: PMC7882955 DOI: 10.1111/jcmm.16192] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Revised: 11/09/2020] [Accepted: 11/17/2020] [Indexed: 12/20/2022] Open
Abstract
microRNA-126 (miR-126), an endothelial-specific miRNA, is associated with vascular homeostasis and angiogenesis. However, the efficiency of miR-126-based treatment is partially compromised due to the low efficiency of miRNA delivery in vivo. Lately, exosomes have emerged as a natural tool for therapeutic molecule delivery. Herein, we investigated whether exosomes derived from bone marrow mesenchymal stem cells (BMMSCs) can be utilized to deliver miR-126 to promote angiogenesis. Exosomes were isolated from BMMSCs overexpressed with miR-126 (Exo-miR-126) by ultracentrifugation. In vitro study, Exo-miR-126 treatment promoted the proliferation, migration and angiogenesis of human umbilical vein endothelial cells (HUVECs). Furthermore, the gene/protein expression of angiogenesis-related vascular endothelial growth factor (VEGF) and angiotensin-1 (Ang-1) were up-regulated after incubation with Exo-miR-126. Additionally, the expression level of phosphoinositol-3 kinase regulatory subunit 2 (PIK3R2) showed an inverse correlation with miR-126 in HUVECs. Particularly, the Exo-miR-126 treatment contributed to enhanced angiogenesis of HUVECs by targeting PIK3R2 to activate the PI3K/Akt signalling pathway. Similarly, Exo-miR-126 administration profoundly increased the number of newly formed capillaries in wound sites and accelerated the wound healing in vivo. The results demonstrate that exosomes derived from BMMSCs combined with miR-126 may be a promising strategy to promote angiogenesis.
Collapse
Affiliation(s)
- Lei Zhang
- Department of Orthopaedic SurgerySecond Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxi ProvinceChina
- Department of Orthopaedic SurgeryXi'an Children's HospitalXi’anShaanxi ProvinceChina
| | - Pengrong Ouyang
- Department of Orthopaedic SurgerySecond Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxi ProvinceChina
| | - Gaole He
- Department of Spine SurgeryHong Hui HospitalXi’an Jiaotong UniversityXi’anShaanxi ProvinceChina
| | - Xiaowei Wang
- Department of Orthopaedic SurgeryXi'an Children's HospitalXi’anShaanxi ProvinceChina
| | - Defu Song
- Department of Orthopaedic SurgeryXi'an Children's HospitalXi’anShaanxi ProvinceChina
| | - Yijun Yang
- Department of Orthopaedic SurgeryXi'an Children's HospitalXi’anShaanxi ProvinceChina
| | - Xijing He
- Department of Orthopaedic SurgerySecond Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxi ProvinceChina
- Xi'an International Medical Center HospitalXi’anShaanxi ProvinceChina
| |
Collapse
|
53
|
Agarwal T, Kazemi S, Costantini M, Perfeito F, Correia CR, Gaspar V, Montazeri L, De Maria C, Mano JF, Vosough M, Makvandi P, Maiti TK. Oxygen releasing materials: Towards addressing the hypoxia-related issues in tissue engineering. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 122:111896. [PMID: 33641899 DOI: 10.1016/j.msec.2021.111896] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 01/09/2021] [Accepted: 01/16/2021] [Indexed: 02/07/2023]
Abstract
Manufacturing macroscale cell-laden architectures is one of the biggest challenges faced nowadays in the domain of tissue engineering. Such living constructs, in fact, pose strict requirements for nutrients and oxygen supply that can hardly be addressed through simple diffusion in vitro or without a functional vasculature in vivo. In this context, in the last two decades, a substantial amount of work has been carried out to develop smart materials that could actively provide oxygen-release to contrast local hypoxia in large-size constructs. This review provides an overview of the currently available oxygen-releasing materials and their synthesis and mechanism of action, highlighting their capacities under in vitro tissue cultures and in vivo contexts. Additionally, we also showcase an emerging concept, herein termed as "living materials as releasing systems", which relies on the combination of biomaterials with photosynthetic microorganisms, namely algae, in an "unconventional" attempt to supply the damaged or re-growing tissue with the necessary supply of oxygen. We envision that future advances focusing on tissue microenvironment regulated oxygen-supplying materials would unlock an untapped potential for generating a repertoire of anatomic scale, living constructs with improved cell survival, guided differentiation, and tissue-specific biofunctionality.
Collapse
Affiliation(s)
- Tarun Agarwal
- Department of Biotechnology, Indian Institute of Technology Kharagpur, West Bengal 721302, India
| | - Sara Kazemi
- Department of Cell Engineering, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Marco Costantini
- Institute of Physical Chemistry - Polish Academy of Sciences, Warsaw, Poland
| | - Francisca Perfeito
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Clara R Correia
- Research Center "E. Piaggio", Department of Information Engineering, University of Pisa, Largo Lucio Lazzarino 1, 56122 Pisa, Italy
| | - Vítor Gaspar
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Leila Montazeri
- Department of Cell Engineering, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Carmelo De Maria
- Research Center "E. Piaggio", Department of Information Engineering, University of Pisa, Largo Lucio Lazzarino 1, 56122 Pisa, Italy
| | - João F Mano
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, 3810-193 Aveiro, Portugal.
| | - Massoud Vosough
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran; Department of Regenerative Medicine, Cell Science Research Centre, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| | - Pooyan Makvandi
- Center for MicroBioRobotics (CMBR), Istituto Italiano di Tecnologia, Pisa, Italy
| | - Tapas Kumar Maiti
- Department of Biotechnology, Indian Institute of Technology Kharagpur, West Bengal 721302, India.
| |
Collapse
|
54
|
Lou P, Liu S, Xu X, Pan C, Lu Y, Liu J. Extracellular vesicle-based therapeutics for the regeneration of chronic wounds: current knowledge and future perspectives. Acta Biomater 2021; 119:42-56. [PMID: 33161186 DOI: 10.1016/j.actbio.2020.11.001] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 10/20/2020] [Accepted: 11/02/2020] [Indexed: 02/07/2023]
Abstract
Chronic wounds are still an intractable medical problem for both clinicians and researchers and cause a substantial social and medical burden. Current clinical approaches can only manage wounds but have limited capacity to promote the regeneration of chronic wounds. As a type of natural nanovesicle, extracellular vesicles (EVs) from multiple cell types (e.g., stem cells, immune cells, and skin cells) have been shown to participate in all stages of skin wound healing including inflammation, proliferation, and remodeling, and display beneficial roles in promoting wound repair. Moreover, EVs can be further re-engineered with genetic/chemical or scaffold material-based strategies for enhanced skin regeneration. In this review, we provide an overview of EV biology and discuss the current findings regarding the roles of EVs in chronic wound healing, particularly in immune regulation, cell proliferation and migration, angiogenesis, and extracellular matrix remodeling, as well as the therapeutic effects of EVs on chronic wounds by genetic modification, in combination with functionalized biomaterials, and as drug carriers. We also discuss the challenges and perspectives of translating EV-based therapies into clinical wound care in the future.
Collapse
|
55
|
Dinç E, Dursun Ö, Yilmaz G, Kurt AH, Ayaz L, Vatansever M, Özer Ö, Yilmaz ŞN. Evaluation of Anti-Inflammatory and Antiapoptotic Effects of Bone Marrow and Adipose-Derived Mesenchymal Stem Cells in Acute Alkaline Corneal Burn. J Ocul Pharmacol Ther 2021; 37:24-34. [PMID: 33275515 DOI: 10.1089/jop.2020.0103] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Purpose: The aim of the present study is to comparatively evaluate the anti-inflammatory and antiapoptotic effects of bone marrow and adipose-derived mesenchymal stem cells (MSCs) applied subconjunctivally after alkaline corneal burn. Methods: Thirty-two rats were divided into 4 groups and included in the study (n = 8). While no intervention was made in the control group, a chemical burn was created by applying 4 μL of NaOH soaked in 6 mm filter paper to the right eye of each subject in the other groups under general anesthesia. While only subconjunctival 0.1 mL phosphate-buffered saline (PBS) was injected to in the group 1, 2 × 106 adipose or bone marrow-derived MSC in 0.1 mL PBS was applied subconjunctivally to the subjects in the remaining groups (Group 2 and 3, respectively). Tissue samples were collected for histological analysis on the third day after the burn. Tissue samples were evaluated light microscopically and immunohistochemically stained for interleukin-1 beta (IL-1β), tumor necrosis factor alpha (TNF-α), caspase-3 (Cas-3), and CD68. Results: The IL-1β and TNF-α staining scores and the number of CD68- and Cas-3-positive stained cells were significantly lower in the groups given bone marrow and adipose-derived MSC compared to the alkaline burn group (P < 0.0001, for all parameters). Epithelial IL-1β and TNF-α staining scores were significantly lower in the bone marrow-derived MSC group compared to the adipose-derived MSC group (P < 0.0001, for all parameters). Conclusions: The presented study shows that both bone-marrow and adipose-derived MSCs support wound healing in the corneal tissue and strongly suppress the inflammation occured in the tissue.
Collapse
Affiliation(s)
- Erdem Dinç
- Department of Ophthalmology and Faculty of Medicine, Mersin University, Mersin, Turkey
| | - Özer Dursun
- Department of Ophthalmology and Faculty of Medicine, Mersin University, Mersin, Turkey
| | - Gülsen Yilmaz
- Department of Histology & Embryology, Faculty of Medicine, Mersin University, Mersin, Turkey
| | - A Hakan Kurt
- Department of Pharmacology, Bolu İzzet Baysal University, Bolu, Turkey
| | - Lokman Ayaz
- Department of Biochemistry, Faculty of Pharmacy, Trakya University, Edirne, Turkey
| | - Mustafa Vatansever
- Department of Ophthalmology and Faculty of Medicine, Mersin University, Mersin, Turkey
| | - Ömer Özer
- Department of Ophthalmology and Faculty of Medicine, Mersin University, Mersin, Turkey
| | - Şakir Necat Yilmaz
- Department of Histology & Embryology, Faculty of Medicine, Mersin University, Mersin, Turkey
| |
Collapse
|
56
|
Khan AQ, Akhtar S, Prabhu KS, Zarif L, Khan R, Alam M, Buddenkotte J, Ahmad A, Steinhoff M, Uddin S. Exosomes: Emerging Diagnostic and Therapeutic Targets in Cutaneous Diseases. Int J Mol Sci 2020; 21:9264. [PMID: 33291683 PMCID: PMC7730213 DOI: 10.3390/ijms21239264] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Revised: 11/28/2020] [Accepted: 12/02/2020] [Indexed: 02/06/2023] Open
Abstract
Skin is the largest human organ and is continuously exposed to various exogenous and endogenous trigger factors affecting body homeostasis. A number of mechanisms, including genetic, inflammatory and autoimmune ones, have been implicated in the pathogenesis of cutaneous diseases. Recently, there has been considerable interest in the role that extracellular vesicles, particularly exosomes, play in human diseases, through their modulation of multiple signaling pathways. Exosomes are nano-sized vesicles secreted by all cell types. They function as cargo carriers shuttling proteins, nucleic acids, lipids etc., thus impacting the cell-cell communications and transfer of vital information/moieties critical for skin homeostasis and disease pathogenesis. This review summarizes the available knowledge on how exosomes affect pathogenesis of cutaneous diseases, and highlights their potential as future targets for the therapy of various skin diseases.
Collapse
Affiliation(s)
- Abdul Q. Khan
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar; (A.Q.K.); (K.S.P.); (M.A.); (J.B.)
| | - Sabah Akhtar
- Department of Biological and Environmental Sciences, Qatar University, Doha 2713, Qatar; (S.A.); (L.Z.)
| | - Kirti S. Prabhu
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar; (A.Q.K.); (K.S.P.); (M.A.); (J.B.)
| | - Lubna Zarif
- Department of Biological and Environmental Sciences, Qatar University, Doha 2713, Qatar; (S.A.); (L.Z.)
| | - Rehan Khan
- Department of Nano-Therapeutics, Institute of Nano Science and Technology, Habitat Centre, Phase 10, Sector 64, Mohali, Punjab 160062, India;
| | - Majid Alam
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar; (A.Q.K.); (K.S.P.); (M.A.); (J.B.)
- Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar
- Department of Dermatology and Venereology, Rumailah Hospital, Hamad Medical Corporation, Doha 3050, Qatar
| | - Joerg Buddenkotte
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar; (A.Q.K.); (K.S.P.); (M.A.); (J.B.)
- Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar
- Department of Dermatology and Venereology, Rumailah Hospital, Hamad Medical Corporation, Doha 3050, Qatar
| | - Aamir Ahmad
- Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Martin Steinhoff
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar; (A.Q.K.); (K.S.P.); (M.A.); (J.B.)
- Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar
- Department of Dermatology and Venereology, Rumailah Hospital, Hamad Medical Corporation, Doha 3050, Qatar
- Department of Medicine, Weill Cornell Medicine Qatar, Qatar Foundation-Education City, Doha 24144, Qatar
- Department of Medicine, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA
- College of Medicine, Qatar University, Doha 2713, Qatar
| | - Shahab Uddin
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar; (A.Q.K.); (K.S.P.); (M.A.); (J.B.)
- Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar
- Department of Dermatology and Venereology, Rumailah Hospital, Hamad Medical Corporation, Doha 3050, Qatar
| |
Collapse
|
57
|
Shi Z, Wang Q, Jiang D. The preventative effect of bone marrow-derived mesenchymal stem cell exosomes on urethral stricture in rats. Transl Androl Urol 2020; 9:2071-2081. [PMID: 33209670 PMCID: PMC7658129 DOI: 10.21037/tau-20-833] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Background Urethral stricture (US) is a major challenge in urology and there is an urgent need for effective therapies for its treatment. Exosomes derived from bone marrow mesenchymal stem cells (BMSCs-Exos) have been shown to be effective in preventing scar and fibrosis formation after tissue injury. However, the potential utility of BMSCs-Exos in the prevention of US remains unknown. We hypothesized that local administration of BMSCs-Exos may influence urethral healing and scar formation in a rat model of US. Methods A previously established model of rat US was used in this study. Sprague Dawley rats were randomly assigned into sham, US, and US + BMSCs-Exos groups. Micro-ultrasound assessment, histopathology, immunohistochemistry, and gene expression analysis were performed at four weeks post-surgery. Results US rats exhibited thick urethral walls with a narrowed lumen, when compared with sham rats. However, these changes were suppressed in the US + BMSCs-Exos group. The preventative effects of BMSCs-Exos on US formation were also apparent histologically. US + BMSCs-Exos rats demonstrated decreased expression of several fibrosis-related genes in urethral tissues, including Col I, fibronectin, and elastin, when compared with US rats. BMSCs-Exos treatment also led to an increase in the expression of angiogenesis-related genes in these tissues, including VEGF, eNOS, and bFGF. Conclusions Our findings therefore demonstrate that the local administration of BMSCs-Exos prevents urethral stricture formation by regulating fibrosis and angiogenesis. These findings provide a basis for an innovative strategy involving the clinical application of exosomes to counteract US formation.
Collapse
Affiliation(s)
- Zhengzhou Shi
- Department of Urology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qi Wang
- Department of Urology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Dapeng Jiang
- Department of Urology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
58
|
Duan M, Zhang Y, Zhang H, Meng Y, Qian M, Zhang G. Epidermal stem cell-derived exosomes promote skin regeneration by downregulating transforming growth factor-β1 in wound healing. Stem Cell Res Ther 2020; 11:452. [PMID: 33097078 PMCID: PMC7584097 DOI: 10.1186/s13287-020-01971-6] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 10/11/2020] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Scar formation, which may be caused by myofibroblast aggregations, is the greatest challenge during skin wound healing in the clinical setting. Studies have indicated that epidermal stem cells (EPSC) improve wound healing and reduce scar formation. METHODS We investigated the therapeutic effects of EPSC-derived exosomes (EPSC-Exos) on skin wound healing in a skin-defect rat model. We also examined the roles of EPSC-Exos-specific microRNAs in inhibiting the differentiation of human dermal fibroblasts (HDF) into myofibroblasts. RESULTS We found that EPSC-Exos increased the wound healing rate and reduced scar formation in rats. Also, EPSC-Exos improved the regeneration levels of skin appendages, nerves and vessels, as well as the natural distribution of collagen. Furthermore, we found these functions may be achieved by inhibiting the activity of transforming growth factor-β1 (TGF-β1) and its downstream genes. The results showed that some specific microRNAs, including miR-16, let-7a, miR-425-5p and miR-142-3p, were enriched in EPSC-Exos. EPSC-Exos-specific microRNAs, especially miR-425-5p and miR-142-3p, played vital roles in inhibiting myofibroblast differentiation via reducing the TGF-β1 expression in dermal fibroblasts. CONCLUSION We found a novel function of EPSC-Exos-specific microRNAs, suggesting that EPSC-Exos might represent a strategy to prevent scar formation during wound healing in the clinical setting.
Collapse
Affiliation(s)
- Mengna Duan
- Department of Prosthodontics, Hospital of Stomatology, Jilin University, 1500 Qinghua Rd., Changchun, 130021, Jilin, China
- Jilin Provincial Laboratory of Biomedical Engineering, Jilin University, 1500 Qinghua Rd., Changchun, 130021, Jilin, China
| | - Yan Zhang
- Department of Prosthodontics, Hospital of Stomatology, Jilin University, 1500 Qinghua Rd., Changchun, 130021, Jilin, China
| | - Haiyang Zhang
- Department of Prosthodontics, Hospital of Stomatology, Jilin University, 1500 Qinghua Rd., Changchun, 130021, Jilin, China
| | - Yupeng Meng
- Department of Prosthodontics, Hospital of Stomatology, Jilin University, 1500 Qinghua Rd., Changchun, 130021, Jilin, China
| | - Ming Qian
- Department of Prosthodontics, Hospital of Stomatology, Jilin University, 1500 Qinghua Rd., Changchun, 130021, Jilin, China.
| | - Guokun Zhang
- Institute of Antler Science and Product Technology, Changchun Sci-Tech University, 1345 Pudong Rd., Changchun, 130600, Jilin, China.
- Institute of Special Animal and Plant Sciences, Chinese Academy of Agricultural Sciences (CAAS), 4899 Juye St., Changchun, 130112, Jilin, China.
| |
Collapse
|
59
|
Parhizkar Roudsari P, Alavi-Moghadam S, Payab M, Sayahpour FA, Aghayan HR, Goodarzi P, Mohamadi-jahani F, Larijani B, Arjmand B. Auxiliary role of mesenchymal stem cells as regenerative medicine soldiers to attenuate inflammatory processes of severe acute respiratory infections caused by COVID-19. Cell Tissue Bank 2020; 21:405-425. [PMID: 32588163 PMCID: PMC7315014 DOI: 10.1007/s10561-020-09842-3] [Citation(s) in RCA: 20] [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: 04/17/2020] [Accepted: 06/13/2020] [Indexed: 02/07/2023]
Abstract
Acute respiratory infections as one of the most common problems of healthcare systems also can be considered as an important reason for worldwide morbidity and mortality from infectious diseases. Coronaviruses are a group of well-known respiratory viruses that can cause acute respiratory infections. At the current state, the 2019 novel coronavirus is cited as the most worldwide problematic agent for the respiratory system. According to investigations, people with old age and underlying diseases are at higher risk of 2019 novel coronavirus infection. Indeed, they may show a severe form of the disease (with severe acute respiratory infections). Based on the promising role of cell therapy and regenerative medicine approaches in the treatment of several life-threatening diseases, it seems that applying cell-based approaches can also be a hopeful strategy for improving subjects with severe acute respiratory infections caused by the 2019 novel coronavirus. Herein, due to the amazing effects of mesenchymal stem cells in the treatment of various diseases, this review focuses on the auxiliary role of mesenchymal stem cells to reduce inflammatory processes of acute respiratory infections caused by the 2019 novel coronavirus.
Collapse
Affiliation(s)
- Peyvand Parhizkar Roudsari
- Metabolomics and Genomics Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Sepideh Alavi-Moghadam
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Moloud Payab
- Obesity and Eating Habits Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Forough Azam Sayahpour
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Hamid Reza Aghayan
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Parisa Goodarzi
- Brain and Spinal Cord Injury Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Fereshteh Mohamadi-jahani
- Brain and Spinal Cord Injury Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Bagher Larijani
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Babak Arjmand
- Metabolomics and Genomics Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
60
|
Maqsood M, Kang M, Wu X, Chen J, Teng L, Qiu L. Adult mesenchymal stem cells and their exosomes: Sources, characteristics, and application in regenerative medicine. Life Sci 2020; 256:118002. [PMID: 32585248 DOI: 10.1016/j.lfs.2020.118002] [Citation(s) in RCA: 118] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Revised: 06/11/2020] [Accepted: 06/20/2020] [Indexed: 02/08/2023]
Abstract
Human mesenchymal stem cells (MSCs) have become a hot topic in the development of cell therapies and bioengineering. All kinds of MSCs are genomic stable and have the self-renewal ability. Main sources of MSCs are bone marrow, adipose tissues, umbilical cord and placental tissues. MSCs can be cultured in many undifferentiated passages to grow into more specialized cells, produce secretory factors and also support trophic functions in the body. Exosomes, derived from MSCs also have great potential in regenerative medicine and tissue engineering. Exosomes are secreted by MSCs and have the same therapeutic potential as their parent cells. MSCs and their exosomes combined with biomaterials can also be more effective in promoting the regeneration of tissues and organs. However, for use of MSCs-exosomes as a clinical agent different MSCs-exosomes have been manufactured and their therapeutics effects demonstrated in clinical studies. But there are still many characteristics which are unknown and many barriers still need to be conquered. In this review, we not only highlighted the characteristics of human MSCs and their exosomes, but also provided their latest therapeutic strategies in regenerative medicine.
Collapse
Affiliation(s)
- Maria Maqsood
- School of Pharmaceutical Sciences, Jiangnan University, Wuxi 214122, China
| | - Mingzhu Kang
- School of Pharmaceutical Sciences, Jiangnan University, Wuxi 214122, China
| | - Xiaotao Wu
- School of Pharmaceutical Sciences, Jiangnan University, Wuxi 214122, China
| | - Jinghua Chen
- School of Pharmaceutical Sciences, Jiangnan University, Wuxi 214122, China
| | - Liping Teng
- Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China.
| | - Lipeng Qiu
- School of Pharmaceutical Sciences, Jiangnan University, Wuxi 214122, China.
| |
Collapse
|
61
|
Zhang Y, Han F, Gu L, Ji P, Yang X, Liu M, Tao K, Hu D. Adipose mesenchymal stem cell exosomes promote wound healing through accelerated keratinocyte migration and proliferation by activating the AKT/HIF-1α axis. J Mol Histol 2020; 51:375-383. [PMID: 32556903 DOI: 10.1007/s10735-020-09887-4] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 06/13/2020] [Indexed: 12/16/2022]
Abstract
Accelerating wound healing is a key consideration for surgeons. The three stages of wound healing include the inflammatory response, cell proliferation and tissue repair, and much research has focused on the migration and proliferation of epidermal cells, since this is one of the most important steps in wound healing. Studies have shown that adipose mesenchymal stem cells (ADSCs) can promote wound healing by releasing exosomes, although the specific mechanism remains unclear. To clarify the role of adipose mesenchymal stem cell exosomes (ADSCs-exo), we constructed a HaCaT cells model and a mouse wound healing model to examine the effects of ADSCs-exo on wound healing. CCK8 assays and the scratch test showed that ADSCs-exo could promote the proliferation and migration of HaCaT cells. Western blotting and real-time PCR showed that ADSCs-exo upregulated the phosphorylation of AKT and the expression of HIF-1α in HaCaT cells. HIF-1α expression was reduced by inhibiting AKT phosphorylation,and the migration of HaCaT cells simultaneously slowed. These results were also confirmed in vivo. In conclusion, we confirmed that ADSCs-exo promote the proliferation and migration of HaCaT cells by regulating the activation of the AKT/HIF-1α signaling pathway, thus promoting wound healing.
Collapse
Affiliation(s)
- Yue Zhang
- Department of Burns and Cutaneous Surgery, Xijing Hospital, The Fourth Military Medical University, 127 Changle West Road, Xi'an, Shaanxi, 710032, China
| | - Fei Han
- Department of Burns and Cutaneous Surgery, Xijing Hospital, The Fourth Military Medical University, 127 Changle West Road, Xi'an, Shaanxi, 710032, China
| | - Lan Gu
- Department of Burns and Cutaneous Surgery, Xijing Hospital, The Fourth Military Medical University, 127 Changle West Road, Xi'an, Shaanxi, 710032, China
| | - Peng Ji
- Department of Burns and Cutaneous Surgery, Xijing Hospital, The Fourth Military Medical University, 127 Changle West Road, Xi'an, Shaanxi, 710032, China
| | - Xuekang Yang
- Department of Burns and Cutaneous Surgery, Xijing Hospital, The Fourth Military Medical University, 127 Changle West Road, Xi'an, Shaanxi, 710032, China
| | - Mengdong Liu
- Department of Burns and Cutaneous Surgery, Xijing Hospital, The Fourth Military Medical University, 127 Changle West Road, Xi'an, Shaanxi, 710032, China
| | - Ke Tao
- Department of Burns and Cutaneous Surgery, Xijing Hospital, The Fourth Military Medical University, 127 Changle West Road, Xi'an, Shaanxi, 710032, China.
| | - Dahai Hu
- Department of Burns and Cutaneous Surgery, Xijing Hospital, The Fourth Military Medical University, 127 Changle West Road, Xi'an, Shaanxi, 710032, China.
| |
Collapse
|
62
|
Luck J, Weil BD, Lowdell M, Mosahebi A. Adipose-Derived Stem Cells for Regenerative Wound Healing Applications: Understanding the Clinical and Regulatory Environment. Aesthet Surg J 2020; 40:784-799. [PMID: 31406975 DOI: 10.1093/asj/sjz214] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
There is growing interest in the regenerative potential of adipose-derived stem cells (ADSCs) for wound healing applications. ADSCs have been shown to promote revascularization, activate local stem cell niches, reduce oxidative stress, and modulate immune responses. Combined with the fact that they can be harvested in large numbers with minimal donor site morbidity, ADSC products represent promising regenerative cell therapies. This article provides a detailed description of the defining characteristics and therapeutic potential of ADSCs, with a focus on understanding how ADSCs promote tissue regeneration and repair. It summarizes the current regulatory environment governing the use of ADSC products across Europe and the United States and examines how various adipose-derived products conform to the current UK legislative framework. Advice is given to clinicians and researchers on how novel ADSC therapeutics may be developed in accordance with regulatory guidelines.
Collapse
Affiliation(s)
| | - Benjamin D Weil
- Centre for Cell, Gene and Tissue Therapeutics, Royal Free Hospital, London, UK
| | | | | |
Collapse
|
63
|
Baradaran-Rafii A, Sarvari M, Alavi-Moghadam S, Payab M, Goodarzi P, Aghayan HR, Larijani B, Rezaei-Tavirani M, Biglar M, Arjmand B. Cell-based approaches towards treating age-related macular degeneration. Cell Tissue Bank 2020; 21:339-347. [PMID: 32157501 DOI: 10.1007/s10561-020-09826-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Accepted: 03/05/2020] [Indexed: 12/19/2022]
Abstract
Age-related macular degeneration as one of the most common causes of worldwide vision loss needs a proper approach for treatment. Therein, cell therapy and regenerative medicine can hold a great promise to be an effective approach. Accordingly, some preclinical and clinical studies were conducted to search around the therapeutic influence of stem cells in Age-related macular degeneration models and subjects. Hereupon, the purpose of the current review is to discuss the mechanisms of age-related macular degeneration, appropriate animal models along with suitable dosage and route of stem cell administration for its treatment.
Collapse
Affiliation(s)
- Alireza Baradaran-Rafii
- Ocular Tissue Engineering Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Ophthalmic Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Masoumeh Sarvari
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Sepideh Alavi-Moghadam
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Moloud Payab
- Obesity and Eating Habits Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Parisa Goodarzi
- Brain and Spinal Cord Injury Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Hamid Reza Aghayan
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Bagher Larijani
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Mahmood Biglar
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Babak Arjmand
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran.
- Metabolomics and Genomics Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
64
|
Paracrine Mechanisms of Mesenchymal Stromal Cells in Angiogenesis. Stem Cells Int 2020; 2020:4356359. [PMID: 32215017 PMCID: PMC7085399 DOI: 10.1155/2020/4356359] [Citation(s) in RCA: 157] [Impact Index Per Article: 31.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Accepted: 02/05/2020] [Indexed: 12/11/2022] Open
Abstract
The role of the mesenchymal stromal cell- (MSC-) derived secretome is becoming increasingly intriguing from a clinical perspective due to its ability to stimulate endogenous tissue repair processes as well as its effective regulation of the immune system, mimicking the therapeutic effects produced by the MSCs. The secretome is a composite product secreted by MSC in vitro (in conditioned medium) and in vivo (in the extracellular milieu), consisting of a protein soluble fraction (mostly growth factors and cytokines) and a vesicular component, extracellular vesicles (EVs), which transfer proteins, lipids, and genetic material. MSC-derived secretome differs based on the tissue from which the MSCs are isolated and under specific conditions (e.g., preconditioning or priming) suggesting that clinical applications should be tailored by choosing the tissue of origin and a priming regimen to specifically correct a given pathology. MSC-derived secretome mediates beneficial angiogenic effects in a variety of tissue injury-related diseases. This supports the current effort to develop cell-free therapeutic products that bring both clinical benefits (reduced immunogenicity, persistence in vivo, and no genotoxicity associated with long-term cell cultures) and manufacturing advantages (reduced costs, availability of large quantities of off-the-shelf products, and lower regulatory burden). In the present review, we aim to give a comprehensive picture of the numerous components of the secretome produced by MSCs derived from the most common tissue sources for clinical use (e.g., AT, BM, and CB). We focus on the factors involved in the complex regulation of angiogenic processes.
Collapse
|
65
|
Masalova OV, Lesnova EI, Klimova RR, Momotyuk ED, Kozlov VV, Ivanova AM, Payushina OV, Butorina NN, Zakirova NF, Narovlyansky AN, Pronin AV, Ivanov AV, Kushch AA. Genetically Modified Mouse Mesenchymal Stem Cells Expressing Non-Structural Proteins of Hepatitis C Virus Induce Effective Immune Response. Vaccines (Basel) 2020; 8:62. [PMID: 32024236 PMCID: PMC7158691 DOI: 10.3390/vaccines8010062] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 01/29/2020] [Accepted: 01/31/2020] [Indexed: 02/08/2023] Open
Abstract
Hepatitis C virus (HCV) is one of the major causes of chronic liver disease and leads to cirrhosis and hepatocarcinoma. Despite extensive research, there is still no vaccine against HCV. In order to induce an immune response in DBA/2J mice against HCV, we obtained modified mouse mesenchymal stem cells (mMSCs) simultaneously expressing five nonstructural HCV proteins (NS3-NS5B). The innate immune response to mMSCs was higher than to DNA immunization, with plasmid encoding the same proteins, and to naïve unmodified MSCs. mMSCs triggered strong phagocytic activity, enhanced lymphocyte proliferation, and production of type I and II interferons. The adaptive immune response to mMSCs was also more pronounced than in the case of DNA immunization, as exemplified by a fourfold stronger stimulation of lymphocyte proliferation in response to HCV, a 2.6-fold higher rate of biosynthesis, and a 30-fold higher rate of secretion of IFN-γ, as well as by a 40-fold stronger production of IgG2a antibodies to viral proteins. The immunostimulatory effect of mMSCs was associated with pronounced IL-6 secretion and reduction in the population of myeloid derived suppressor cells (MDSCs). Thus, this is the first example that suggests the feasibility of using mMSCs for the development of an effective anti-HCV vaccine.
Collapse
Affiliation(s)
- Olga V. Masalova
- Gamaleya National Research Center of Epidemiology and Microbiology, Ministry of Health of the Russian Federation, Moscow 123098, Russia; (E.I.L.); (R.R.K.); (E.D.M.); (V.V.K.); (A.M.I.); (A.N.N.); (A.V.P.); (A.A.K.)
| | - Ekaterina I. Lesnova
- Gamaleya National Research Center of Epidemiology and Microbiology, Ministry of Health of the Russian Federation, Moscow 123098, Russia; (E.I.L.); (R.R.K.); (E.D.M.); (V.V.K.); (A.M.I.); (A.N.N.); (A.V.P.); (A.A.K.)
| | - Regina R. Klimova
- Gamaleya National Research Center of Epidemiology and Microbiology, Ministry of Health of the Russian Federation, Moscow 123098, Russia; (E.I.L.); (R.R.K.); (E.D.M.); (V.V.K.); (A.M.I.); (A.N.N.); (A.V.P.); (A.A.K.)
| | - Ekaterina D. Momotyuk
- Gamaleya National Research Center of Epidemiology and Microbiology, Ministry of Health of the Russian Federation, Moscow 123098, Russia; (E.I.L.); (R.R.K.); (E.D.M.); (V.V.K.); (A.M.I.); (A.N.N.); (A.V.P.); (A.A.K.)
| | - Vyacheslav V. Kozlov
- Gamaleya National Research Center of Epidemiology and Microbiology, Ministry of Health of the Russian Federation, Moscow 123098, Russia; (E.I.L.); (R.R.K.); (E.D.M.); (V.V.K.); (A.M.I.); (A.N.N.); (A.V.P.); (A.A.K.)
| | - Alla M. Ivanova
- Gamaleya National Research Center of Epidemiology and Microbiology, Ministry of Health of the Russian Federation, Moscow 123098, Russia; (E.I.L.); (R.R.K.); (E.D.M.); (V.V.K.); (A.M.I.); (A.N.N.); (A.V.P.); (A.A.K.)
| | - Olga V. Payushina
- Federal State Autonomous Educational Institution of Higher Education I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), Moscow 119991, Russia;
| | - Nina N. Butorina
- Koltzov Institute of Developmental Biology of Russian Academy of Sciences, Moscow 119334, Russia;
| | - Natalia F. Zakirova
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow 119991, Russia;
| | - Alexander N. Narovlyansky
- Gamaleya National Research Center of Epidemiology and Microbiology, Ministry of Health of the Russian Federation, Moscow 123098, Russia; (E.I.L.); (R.R.K.); (E.D.M.); (V.V.K.); (A.M.I.); (A.N.N.); (A.V.P.); (A.A.K.)
| | - Alexander V. Pronin
- Gamaleya National Research Center of Epidemiology and Microbiology, Ministry of Health of the Russian Federation, Moscow 123098, Russia; (E.I.L.); (R.R.K.); (E.D.M.); (V.V.K.); (A.M.I.); (A.N.N.); (A.V.P.); (A.A.K.)
| | - Alexander V. Ivanov
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow 119991, Russia;
| | - Alla A. Kushch
- Gamaleya National Research Center of Epidemiology and Microbiology, Ministry of Health of the Russian Federation, Moscow 123098, Russia; (E.I.L.); (R.R.K.); (E.D.M.); (V.V.K.); (A.M.I.); (A.N.N.); (A.V.P.); (A.A.K.)
| |
Collapse
|
66
|
Ebrahimi-Barough S, Ai J, Payab M, Alavi-Moghadam S, Shokati A, Aghayan HR, Larijani B, Arjmand B. Standard Operating Procedure for the Good Manufacturing Practice-Compliant Production of Human Endometrial Stem Cells for Multiple Sclerosis. Methods Mol Biol 2020; 2286:199-212. [PMID: 32504294 DOI: 10.1007/7651_2020_281] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Multiple sclerosis (MS) is the most common cause of neurological diseases. Although, there are some effective medications with regulatory approval for treating MS, they are only partially effective and cannot promote repairing of tissue damage directly which occurs in the central nervous system. Therefore, there is an essential need to develop novel therapeutic approaches for neuroprotection or repairing damaged tissue in MS. Accordingly, cell-based therapies as a novel therapeutic strategy have opened a new horizon in treatment of MS. Each setting in cell therapy has potential benefits. Human endometrial stem cells as an invaluable source for cell therapy have introduced treatment for MS. In this respect, good manufacturing practice (GMP) has a pivotal role in clinical production of stem cells. This chapter tries to describe the protocol of GMP-grade endometrial stem cells for treatment of MS.
Collapse
Affiliation(s)
- Somayeh Ebrahimi-Barough
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Jafar Ai
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Moloud Payab
- Obesity and Eating Habits Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Sepideh Alavi-Moghadam
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Ameneh Shokati
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Hamid Reza Aghayan
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Bagher Larijani
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Babak Arjmand
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran. .,Metabolomics and Genomics Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
67
|
Huang C, Luo WF, Ye YF, Lin L, Wang Z, Luo MH, Song QD, He XP, Chen HW, Kong Y, Tang YK. Characterization of inflammatory factor-induced changes in mesenchymal stem cell exosomes and sequencing analysis of exosomal microRNAs. World J Stem Cells 2019; 11:859-890. [PMID: 31692888 PMCID: PMC6828590 DOI: 10.4252/wjsc.v11.i10.859] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 03/24/2019] [Accepted: 07/30/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Treatments utilizing stems cells often require stem cells to be exposed to inflammatory environments, but the effects of such environments are unknown. AIM To examine the effects of inflammatory cytokines on the morphology and quantity of mesenchymal stem cell exosomes (MSCs-exo) as well as the differential expression of microRNAs (miRNAs) in the exosomes. METHODS MSCs were isolated from human umbilical tissue by enzymatic digestion. Exosomes were then collected after a 48-h incubation period in a serum-free medium with one of the following the inflammatory cytokines: None (control), vascular cell adhesion molecule-1 (VCAM-1), tumor necrosis factor (TNF) α, and interleukin (IL) 6. The morphology and quantity of each group of MSC exosomes were observed and measured. The miRNAs in MSCs-exo were sequenced. We compared the sequenced data with the miRBase and other non-coding databases in order to detect differentially expressed miRNAs and explore their target genes and regulatory mechanisms. In vitro tube formation assays and Western blot were performed in endothelial cells which were used to assess the angiogenic potential of MSCs-exo after inflammatory cytokine stimulation. RESULTS MSCs-exo were numerous, small, and regularly shaped in the VCAM-1 group. TNFα stimulated MSCs to secrete larger and irregular exosomes. IL6 led to a reduced quantity of MSCs-exo. Compared to the control group, the TNFα and IL6 groups had more downregulated differentially expressed miRNAs, particularly angiogenesis-related miRNAs. The angiogenic potential of MSCs-exo declined after IL6 stimulation. CONCLUSION TNFα and IL6 may influence the expression of miRNAs that down-regulate the PI3K-AKT, MAPK, and VEGF signaling pathways; particularly, IL6 significantly down-regulates the PI3K-AKT signaling pathway. Overall, inflammatory cytokines may lead to changes in exosomal miRNAs that abnormally impact cellular components, molecular function, and biological processes.
Collapse
Affiliation(s)
- Chen Huang
- Department of Minimally Invasive Interventional Radiology, Guangzhou Panyu Central Hospital, Guangzhou 511400, Guangdong Province, China
| | - Wen-Feng Luo
- Department of Minimally Invasive Interventional Radiology, Guangzhou Panyu Central Hospital, Guangzhou 511400, Guangdong Province, China
| | - Yu-Feng Ye
- Department of Minimally Invasive Interventional Radiology, Guangzhou Panyu Central Hospital, Guangzhou 511400, Guangdong Province, China
| | - Li Lin
- Jinan University Biomedical Translational Research Institute, Jinan University, Guangzhou 510632, Guangdong Province, China
| | - Zhe Wang
- Department of Pharmacy, the First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou 510080, Guangdong Province, China
| | - Ming-Hua Luo
- Department of Radiology, Shiyan People's Hospital, Shenzhen 518108, Guangdong Province, China
| | - Qi-De Song
- Department of Minimally Invasive Interventional Radiology, Guangzhou Panyu Central Hospital, Guangzhou 511400, Guangdong Province, China
| | - Xue-Ping He
- Department of Minimally Invasive Interventional Radiology, Guangzhou Panyu Central Hospital, Guangzhou 511400, Guangdong Province, China
| | - Han-Wei Chen
- Department of Minimally Invasive Interventional Radiology, Guangzhou Panyu Central Hospital, Guangzhou 511400, Guangdong Province, China
| | - Yi Kong
- Department of Pharmacy, the First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou 510080, Guangdong Province, China
| | - Yu-Kuan Tang
- Department of Minimally Invasive Interventional Radiology, Guangzhou Panyu Central Hospital, Guangzhou 511400, Guangdong Province, China.
| |
Collapse
|
68
|
Liu K, Chen C, Zhang H, Chen Y, Zhou S. Adipose stem cell‐derived exosomes in combination with hyaluronic acid accelerate wound healing through enhancing re‐epithelialization and vascularization. Br J Dermatol 2019; 181:854-856. [PMID: 30953591 DOI: 10.1111/bjd.17984] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- K. Liu
- Plastic and Reconstructive Surgery Shanghai 9th Peoples Hospital Affiliated to Shanghai Jiaotong University School of Medicine Shanghai P.R. China
| | - C. Chen
- Shanghai Key Laboratory of Tissue Engineering Shanghai P.R. China
| | - H. Zhang
- Shanghai Key Laboratory of Tissue Engineering Shanghai P.R. China
| | - Y. Chen
- Shanghai Key Laboratory of Tissue Engineering Shanghai P.R. China
| | - S. Zhou
- Plastic and Reconstructive Surgery Shanghai 9th Peoples Hospital Affiliated to Shanghai Jiaotong University School of Medicine Shanghai P.R. China
| |
Collapse
|
69
|
Hong P, Yang H, Wu Y, Li K, Tang Z. The functions and clinical application potential of exosomes derived from adipose mesenchymal stem cells: a comprehensive review. Stem Cell Res Ther 2019; 10:242. [PMID: 31391108 PMCID: PMC6686455 DOI: 10.1186/s13287-019-1358-y] [Citation(s) in RCA: 184] [Impact Index Per Article: 30.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Exosomes are extracellular membranous nanovesicles that mediate local and systemic intercellular communication by transporting proteins or nucleic acids (DNA and RNA) into target cells, thus altering the behaviors of recipient cells. Recent studies have revealed that these vesicles play a critical role in many biological functions, such as cell proliferation, immune regulation, nerve regeneration, and cancer. Adipose-derived stem cells (ADSCs) are now considered a multipotent and abundant tool in the field of cell therapy and regenerative medicine. ADSCs can produce and secrete many exosomes, which inherit multiple functions of cells. Therefore, in this review, we will introduce the characteristics of exosomes derived from ADSCs (ADSC-Exos), describe their functions in different biological processes, summarize the latest research achievements, describe their limitations in cell-free therapy, and provide further insights into their clinical application potential for the treatment of certain diseases.
Collapse
Affiliation(s)
- Pengyu Hong
- Department of Oral & Maxillofacial Surgery, Xiangya Stomatological Hospital & School of Stomatology, Central South University, Changsha, 410008 Hunan China
| | - Hao Yang
- Department of Oral & Maxillofacial Surgery, Xiangya Stomatological Hospital & School of Stomatology, Central South University, Changsha, 410008 Hunan China
| | - Yue Wu
- Department of Oral & Maxillofacial Surgery, Xiangya Stomatological Hospital & School of Stomatology, Central South University, Changsha, 410008 Hunan China
| | - Kun Li
- Department of Oral & Maxillofacial Surgery, Xiangya Stomatological Hospital & School of Stomatology, Central South University, Changsha, 410008 Hunan China
| | - Zhangui Tang
- Department of Oral & Maxillofacial Surgery, Xiangya Stomatological Hospital & School of Stomatology, Central South University, Changsha, 410008 Hunan China
| |
Collapse
|
70
|
Yang R, Liu F, Wang J, Chen X, Xie J, Xiong K. Epidermal stem cells in wound healing and their clinical applications. Stem Cell Res Ther 2019; 10:229. [PMID: 31358069 PMCID: PMC6664527 DOI: 10.1186/s13287-019-1312-z] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The skin has important barrier, sensory, and immune functions, contributing to the health and integrity of the organism. Extensive skin injuries that threaten the entire organism require immediate and effective treatment. Wound healing is a natural response, but in severe conditions, such as burns and diabetes, this process is insufficient to achieve effective treatment. Epidermal stem cells (EPSCs) are a multipotent cell type and are committed to the formation and differentiation of the functional epidermis. As the contributions of EPSCs in wound healing and tissue regeneration have been increasingly attracting the attention of researchers, a rising number of therapies based on EPSCs are currently under development. In this paper, we review the characteristics of EPSCs and the mechanisms underlying their functions during wound healing. Applications of EPSCs are also discussed to determine the potential and feasibility of using EPSCs clinically in wound healing.
Collapse
Affiliation(s)
- Ronghua Yang
- Department of Burn Surgery, The First People’s Hospital of Foshan, Foshan, 528000 China
| | - Fengxia Liu
- Department of Human Anatomy, School of Basic Medical Science, Xinjiang Medical University, Urumqi, 830001 China
| | - Jingru Wang
- Department of Burn Surgery, The First People’s Hospital of Foshan, Foshan, 528000 China
| | - Xiaodong Chen
- Department of Burn Surgery, The First People’s Hospital of Foshan, Foshan, 528000 China
| | - Julin Xie
- Department of Burn Surgery, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 512100 China
| | - Kun Xiong
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Morphological Sciences Building, Central South University, 172 Tongzi Po Road, Changsha, 410013 Hunan China
| |
Collapse
|
71
|
Annexin A1 Contained in Extracellular Vesicles Promotes the Activation of Keratinocytes by Mesoglycan Effects: An Autocrine Loop Through FPRs. Cells 2019; 8:cells8070753. [PMID: 31331117 PMCID: PMC6679056 DOI: 10.3390/cells8070753] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 07/09/2019] [Accepted: 07/18/2019] [Indexed: 12/11/2022] Open
Abstract
We have recently demonstrated that mesoglycan, a fibrinolytic compound, may be a promising pro-healing drug for skin wound repair. We showed that mesoglycan induces migration, invasion, early differentiation, and translocation to the membrane of keratinocytes, as well as the secretion of annexin A1 (ANXA1), further involved in keratinocytes activation. These events are triggered by the syndecan-4 (SDC4)/PKCα pathway. SDC4 also participates to the formation and secretion of microvesicles (EVs) which may contribute to wound healing. EVs were isolated from HaCaT cells, as human immortalized keratinocytes, and then characterised by Western blotting, Field Emission-Scanning Electron Microscopy, and Dynamic Light Scattering. Their autocrine effects were investigated by Wound-Healing/invasion assays and confocal microscopy to analyse cell motility and differentiation, respectively. Here, we found that the mesoglycan increased the release of EVs which amplify its same effects. ANXA1 contained in the microvesicles is able to promote keratinocytes motility and differentiation by acting on Formyl Peptide Receptors (FPRs). Thus, the extracellular form of ANXA1 may be considered as a link to intensify the effects of mesoglycan. In this study, for the first time, we have identified an interesting autocrine loop ANXA1/EVs/FPRs in human keratinocytes, induced by mesoglycan.
Collapse
|
72
|
Goodarzi P, Alavi-Moghadam S, Payab M, Larijani B, Rahim F, Gilany K, Bana N, Tayanloo-Beik A, Foroughi Heravani N, Hadavandkhani M, Arjmand B. Metabolomics Analysis of Mesenchymal Stem Cells. INTERNATIONAL JOURNAL OF MOLECULAR AND CELLULAR MEDICINE 2019; 8:30-40. [PMID: 32351907 PMCID: PMC7175611 DOI: 10.22088/ijmcm.bums.8.2.30] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Accepted: 05/20/2019] [Indexed: 12/12/2022]
Abstract
Various mesenchymal stem cells as easily accessible and multipotent cells can share different essential signaling pathways related to their stemness ability. Understanding the mechanism of stemness ability can be useful for controlling the stem cells for regenerative medicine targets. In this context, OMICs studies can analyze the mechanism of different stem cell properties or stemness ability via a broad range of current high-throughput techniques. This field is fundamentally directed toward the analysis of whole genome (genomics), mRNAs (transcriptomics), proteins (proteomics) and metabolites (metabolomics) in biological samples. According to several studies, metabolomics is more effective than other OMICs ّfor various system biology concerns. Metabolomics can elucidate the biological mechanisms of various mesenchymal stem cell function by measuring their metabolites such as their secretome components. Analyzing the metabolic alteration of mesenchymal stem cells can be useful to promote their regenerative medicine application.
Collapse
Affiliation(s)
- Parisa Goodarzi
- Brain and Spinal Cord Injury Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Sepideh Alavi-Moghadam
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Moloud Payab
- Obesity and Eating Habits Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Bagher Larijani
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical sciences, Tehran, Iran
| | - Fakher Rahim
- Health Research Institute, Thalassemia and Hemoglobinopathies Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Kambiz Gilany
- Integrative Oncology Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran .,Department of Biomedical Sciences, University of Antwerp, Belgium
| | - Nikoo Bana
- Metabolomics and Genomics Research Center, Endocrinology and Metabolism Molecular- Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Akram Tayanloo-Beik
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Najmeh Foroughi Heravani
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahdieh Hadavandkhani
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Babak Arjmand
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran .,Metabolomics and Genomics Research Center, Endocrinology and Metabolism Molecular- Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
73
|
Li QC, Liang Y, Su ZB. Prophylactic treatment with MSC-derived exosomes attenuates traumatic acute lung injury in rats. Am J Physiol Lung Cell Mol Physiol 2019; 316:L1107-L1117. [PMID: 30892077 DOI: 10.1152/ajplung.00391.2018] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
The mesenchymal stem cell (MSC) is a potential strategy in the pretreatment of traumatic acute lung injury (ALI), a disease that causes inflammation and oxidative stress. This study aimed to investigate whether MSC-exosomal microRNA-124-3p (miR-124-3p) affects traumatic ALI. Initially, a traumatic ALI rat model was established using the weight-drop method. Then, exosomes were obtained from MSCs of Sprague-Dawley rats, which were injected into the traumatic ALI rats. We found that miR-124-3p was abundantly-expressed in MSCs-derived exosomes and could directly target purinergic receptor P2X ligand-gated ion channel 7 (P2X7), which was overexpressed in traumatic ALI rats. After that, a loss- and gain-of-function study was performed in MSCs and traumatic ALI rats to investigate the role of miR-124-3p and P2X7 in traumatic ALI. MSC-derived exosomal miR-124-3p or silenced P2X7 was observed to increase the survival rate of traumatic ALI rats and enhance the glutathione/superoxide dismutase activity in their lung tissues. However, the wet/dry weight of lung tissues, activity of methylenedioxyamphetamine and H2O2, and levels of inflammatory factors (TNF-a, IL-6, and IL-8) were reduced. Similarly, the numbers of total cells, macrophages, neutrophils, and lymphocytes in bronchoalveolar lavage fluid were also reduced when treated with exosomal miR-124-3p or silenced P2X7. In conclusion, the results provide evidence that miR-124-3p transferred by MSC-derived exosomes inhibited P2X7 expression, thus improving oxidative stress injury and suppressing inflammatory response in traumatic ALI, highlighting a potential pretreatment for traumatic ALI.
Collapse
Affiliation(s)
- Qing-Chun Li
- Department of Gastrointestinal Colorectal and Anal Surgery, China-Japan Union Hospital of Jilin University, Changchun, People’s Republic of China
| | - Yun Liang
- Center of Physical Examination, China-Japan Union Hospital of Jilin University, Changchun, People’s Republic of China
| | - Zhen-Bo Su
- Department of Anesthesiology, China-Japan Union Hospital of Jilin University, Changchun, People’s Republic of China
| |
Collapse
|
74
|
Lee GW, Seo MS, Kang KK, Oh SK. Epidural Fat-Derived Mesenchymal Stem Cell: First Report of Epidural Fat-Derived Mesenchymal Stem Cell. Asian Spine J 2019; 13:361-367. [PMID: 30669827 PMCID: PMC6547395 DOI: 10.31616/asj.2018.0215] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 10/04/2018] [Accepted: 11/03/2018] [Indexed: 12/18/2022] Open
Abstract
STUDY DESIGN Experimental study. PURPOSE To determine whether epidural fat (EF) tissue contains mesenchymal stem cells (MSC). OVERVIEW OF LITERATURE Spine surgeons are unaware of the contents of EF tissue and the reason for its presence between the ligamentum flavum and the dura mater; therefore, EF tissues are routinely eliminated during surgical procedures. However, EF removal causes certain postoperative problems, such as post-laminectomy syndrome. We hypothesized that the EF tissue may play a significant supportive role for the neural structures and other nearby conditions. METHODS EF tissues were obtained from consenting patients (n=3) during posterior decompression surgery of the lumbar spine. The primary cells were isolated and cultured as per previously described methods with some modifications, and the cell morphology and cumulation were examined. Thereafter, reverse transcription-polymerase chain reaction (RT-PCR), a fluorescence-activated cell sorting (FACS) analysis, and differentiation potency for differentiation into osteoblasts, chondroblasts, and adipocytes were investigated to identify whether the cells derived from EF are MSC. RESULTS The cells from the EF tissue had a fibroblast or neuron-like morphology that persisted until the senescence at p18. MSCspecific genes, such as OCT4, SOX2, KLF4, MYC, and GAPDH were expressed in the RT-PCR study, while MSC-specific surface markers such as CD105, CD90, and CD73 were exhibited in the FACS analysis. The differentiation properties of EF-MSC for differentiation into the three types of cells (osteoblast, chondroblast, and adipocyte) were also confirmed. CONCLUSIONS Based on the cell culture, FACS analysis, RT-PCR analysis, and differentiation potent outcomes, all the features of the cells corresponded to MSC. This is the first study to identify EF-MSC derived from the EF tissue.
Collapse
Affiliation(s)
- Gun Woo Lee
- Department of Orthopaedic Surgery, Spine Center, Yeungnam University Medical Center, Yeungnam University College of Medicine, Daegu, Korea
| | - Min-Soo Seo
- Labaratory Animal Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu, Korea
| | - Kyung-Ku Kang
- Labaratory Animal Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu, Korea
| | - Se-Kyung Oh
- Labaratory Animal Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu, Korea
| |
Collapse
|
75
|
Lu Y, Zhou Y, Zhang R, Wen L, Wu K, Li Y, Yao Y, Duan R, Jia Y. Bone Mesenchymal Stem Cell-Derived Extracellular Vesicles Promote Recovery Following Spinal Cord Injury via Improvement of the Integrity of the Blood-Spinal Cord Barrier. Front Neurosci 2019; 13:209. [PMID: 30914918 PMCID: PMC6423165 DOI: 10.3389/fnins.2019.00209] [Citation(s) in RCA: 136] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2018] [Accepted: 02/22/2019] [Indexed: 01/09/2023] Open
Abstract
Mesenchymal stem cell (MSC) transplantation has been shown to represent a potential treatment for traumatic spinal cord injury (SCI). However, there are several obstacles that need to be overcome before MSCs can be considered for clinical application, such as failure of MSCs to reach the spinal cord lesion core and possible tumor formation. Recent studies have suggested that MSC treatment is beneficial owing to paracrine-secreted factors. Extracellular vesicles are considered to be some of the most valuable paracrine molecules. However, the therapeutic mechanism of extracellular vesicles on spinal cord injury has not been studied clearly. Therefore, our study investigated the effect of systemic administration of extracellular vesicles on the loss of motor function after SCI and examined the potential mechanisms underlying their effects. Disruption of the blood-spinal cord barrier (BSCB) is a crucial factor that can be detrimental to motor function recovery. Pericytes are an important component of the neurovascular unit, and play a pivotal role in maintaining the structural integrity of the BSCB. Our study demonstrated that administration of bone mesenchymal stem cell-derived extracellular vesicles (BMSC-EV) reduced brain cell death, enhanced neuronal survival and regeneration, and improved motor function compared with the administration of BMSC-EV free culture media (EV-free CM). Besides, the BSCB was attenuated and pericyte coverage was significantly decreased in vivo. Furthermore, we found that exosomes reduced pericyte migration via downregulation of NF-κB p65 signaling, with a consequent decrease in the permeability of the BSCB. In summary, we identified that extracellular vesicles treatment suppressed the migration of pericytes and further improved the integrity of the BSCB via NF-κB p65 signaling in pericytes. Our data suggest that extracellular vesicles may serve as a promising treatment strategy for SCI.
Collapse
Affiliation(s)
- Yanhui Lu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Yan Zhou
- Department of Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Ruiyi Zhang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Lulu Wen
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Kaimin Wu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yanfei Li
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yaobing Yao
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Ranran Duan
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yanjie Jia
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
76
|
Ma T, Fu B, Yang X, Xiao Y, Pan M. Adipose mesenchymal stem cell-derived exosomes promote cell proliferation, migration, and inhibit cell apoptosis via Wnt/β-catenin signaling in cutaneous wound healing. J Cell Biochem 2019; 120:10847-10854. [PMID: 30681184 DOI: 10.1002/jcb.28376] [Citation(s) in RCA: 190] [Impact Index Per Article: 31.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Accepted: 01/10/2019] [Indexed: 12/15/2022]
Abstract
Cutaneous wounds, a type of soft tissue injury, are difficult to heal in aging. Differentiation, migration, proliferation, and apoptosis of skin cells are identified as key factors during wound healing processes. Mesenchymal stem cells have been documented as possible candidates for wound healing treatment because their use could augment the regenerative capacity of many tissues. However, the effects of exosomes derived from adipose-derived stem cell (ADSC-exos) on cutaneous wound healing remain to be carefully elucidated. In this present study, HaCaT cells were exposed to hydrogen peroxide (H2 O 2 ) for the establishment of the skin lesion model. Cell Counting Kit-8 assay, migration assay, and flow cytometry assay were conducted to detect the biological function of ADSC-exos in skin lesion model. Finally, the possible mechanism was further investigated using Western blot assay. The successful construction of the skin lesion model was confirmed by results of the enhanced cell apoptosis of HaCaT cells induced by H 2 O 2 , the increased Bax expression and decreased Bcl-2 expression. CD9 and CD63 expression evidenced the existence of ADSC-exos. The results of functional experiments demonstrated that ADSC-exos could prompt cell proliferation and migration of HaCaT cells, and repress cell apoptosis of HaCaT cells. In addition, the activation of Wnt/β-catenin signaling was confirmed by the enhanced expression of β-catenin at the protein level. Collectively, our findings suggest that ADSC-exos play a positive role in cutaneous wound healing possibly via Wnt/β-catenin signaling. Our study may provide new insights into the therapeutic target for cutaneous wound healing.
Collapse
Affiliation(s)
- Tao Ma
- Department of Oral and Maxillofacial Surgery, Affiliated Hospital of Guilin Medical University, Guilin, China
| | - Bingchuan Fu
- Department of Medical Cosmetology, Tianyou Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, Hubei, China
| | - Xin Yang
- Department of Otolaryngology, The 161th Hospital of the Chinese People's Liberation Army, Wuhan, Hubei, China
| | - Yilei Xiao
- Department of Neurosurgery, Liaocheng People's Hospital, Liaocheng, Shandong, China
| | - Mengxiong Pan
- Department of Oral and Maxillofacial Surgery, Affiliated Hospital of Guilin Medical University, Guilin, China
| |
Collapse
|
77
|
Novel trends in application of stem cells in skin wound healing. Eur J Pharmacol 2018; 843:307-315. [PMID: 30537490 DOI: 10.1016/j.ejphar.2018.12.012] [Citation(s) in RCA: 114] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 11/28/2018] [Accepted: 12/06/2018] [Indexed: 12/15/2022]
Abstract
The latest findings indicate the huge therapeutic potential of stem cells in regenerative medicine, including the healing of chronic wounds. Main stem cell types involved in wound healing process are: epidermal and dermal stem cells, mesenchymal stem cells (MSCs), endothelial progenitor cells (EPCs) and hematopoietic stem cells (HSCs). In the therapy of chronic wounds, they can be administrated either topically or using different matrix like hydrogels, scaffolds, dermal substitutes and extracellular matrix (ECM) derivatives. Stem cells are proven to positively influence wound healing by different direct and indirect mechanisms including residing cells stimulation, biomolecules release, inflammation control and ECM remodelling. MSCs are especially worth mentioning as they can be easily derived from bone-marrow or adipose tissue. Apart from traditional approach of administering living stem cells to wounds, new trends have emerged in recent years. Good healing results are obtained using stem cell secretome alone, for example exosomes or conditioned media. There are also attempts to improve healing potential of stem cells by their co-culture with other cell types as well as by their genetic modifications or pretreatment using different chemicals or cell media. Moreover, stem cells have been tested for novel therapeutic purposes like for example acute burns and have been used in experiments on large animal models including pigs and sheep. In this review we discuss the role of stem cells in skin wound healing acceleration. In addition, we analyse possible new strategies of stem cells application in treatment of chronic wounds.
Collapse
|