1
|
Nouri S, Shokraneh S, Fatehi Shalamzari P, Ahmed MH, Radi UK, Idan AH, Ebrahimi MJ, Moafi M, Gholizadeh N. Application of Mesenchymal Stem Cells and Exosome alone or Combination Therapy as a Treatment Strategy for Wound Healing. Cell Biochem Biophys 2024; 82:3209-3222. [PMID: 39068609 DOI: 10.1007/s12013-024-01448-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] [Accepted: 07/17/2024] [Indexed: 07/30/2024]
Abstract
The process of wound healing consists of multiple phases, and any disruptions in these phases can lead to the wound becoming chronic and impose heavy financial and psychological costs on the patient and a huge economic burden on the country's healthcare system. Various treatments such as drugs, matrix and scaffolds, blood products, cell therapy, and a combination of these treatments are used for wound healing. The use of mesenchymal stem cells (MSCs) is one of these methods that have produced appropriate responses in the healing of patients' wounds. MSCs by secreting growth factors, cytokines, chemokines, and RNAs elicit changes in cell proliferation, migration, growth, signaling, immunomodulation, and wound re-epithelialization process, and as a result, accelerate wound closure and wound healing. These cells can be isolated from different body sources with different cell characteristics and used directly on the wound site or by injection. In addition, MSCs-derived exosomes have attracted growing attention due to circumventing concerns relating to the direct use of MSCs. To increase the performance of MSCs, they can be used together with other compounds such as platelets, matrices, or scaffolds. This study examined the functions of MSCs in wound healing, as well as the vesicles they secrete, cellular and molecular mechanisms, and combined treatments with MSCs for wound healing.
Collapse
Affiliation(s)
- Soheil Nouri
- Department of Hematology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | | | | | | | - Usama Kadem Radi
- Collage of Pharmacy, National University of Science and Technology, Dhi Qar, 64001, Iraq
| | | | - Mohammad Javad Ebrahimi
- Cell Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Maral Moafi
- Cell Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Nasim Gholizadeh
- Department of Dermatology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran.
| |
Collapse
|
2
|
Li D, Li D, Wang Z, Li J, Shahzad KA, Wang Y, Tan F. Signaling pathways activated and regulated by stem cell-derived exosome therapy. Cell Biosci 2024; 14:105. [PMID: 39164778 PMCID: PMC11334359 DOI: 10.1186/s13578-024-01277-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Accepted: 07/10/2024] [Indexed: 08/22/2024] Open
Abstract
Stem cell-derived exosomes exert comparable therapeutic effects to those of their parental stem cells without causing immunogenic, tumorigenic, and ethical disadvantages. Their therapeutic advantages are manifested in the management of a broad spectrum of diseases, and their dosing versatility are exemplified by systemic administration and local delivery. Furthermore, the activation and regulation of various signaling cascades have provided foundation for the claimed curative effects of exosomal therapy. Unlike other relevant reviews focusing on the upstream aspects (e.g., yield, isolation, modification), and downstream aspects (e.g. phenotypic changes, tissue response, cellular behavior) of stem cell-derived exosome therapy, this unique review endeavors to focus on various affected signaling pathways. After meticulous dissection of relevant literature from the past five years, we present this comprehensive, up-to-date, disease-specific, and pathway-oriented review. Exosomes sourced from various types of stem cells can regulate major signaling pathways (e.g., the PTEN/PI3K/Akt/mTOR, NF-κB, TGF-β, HIF-1α, Wnt, MAPK, JAK-STAT, Hippo, and Notch signaling cascades) and minor pathways during the treatment of numerous diseases encountered in orthopedic surgery, neurosurgery, cardiothoracic surgery, plastic surgery, general surgery, and other specialties. We provide a novel perspective in future exosome research through bridging the gap between signaling pathways and surgical indications when designing further preclinical studies and clinical trials.
Collapse
Affiliation(s)
- Ding Li
- Department of ORL-HNS, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Danni Li
- Department of ORL-HNS, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, China
- Plasma Medicine and Surgical Implants Center, Tongji University, Shanghai, China
| | - Zhao Wang
- Department of ORL-HNS, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jiaojiao Li
- Department of ORL-HNS, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, China
- Plasma Medicine and Surgical Implants Center, Tongji University, Shanghai, China
| | - Khawar Ali Shahzad
- Department of ORL-HNS, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, China
- Plasma Medicine and Surgical Implants Center, Tongji University, Shanghai, China
| | - Yanhong Wang
- Department of ORL-HNS, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, China
- Plasma Medicine and Surgical Implants Center, Tongji University, Shanghai, China
| | - Fei Tan
- Department of ORL-HNS, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, China.
- Plasma Medicine and Surgical Implants Center, Tongji University, Shanghai, China.
- The Royal College of Surgeons in Ireland, Dublin, Ireland.
- The Royal College of Surgeons of England, London, UK.
| |
Collapse
|
3
|
Yang X, Zhang S, Chen K, Shen D, Yang Y, Shen A, Liang J, Xu M, Yang Y, Zhao Y, Li H, Tong X. Hypoxic Preconditioned ADSC Exosomes Enhance Vaginal Wound Healing via Accelerated Keratinocyte Proliferation and Migration Through AKT/HIF‑1α Axis Activation. Cell Mol Bioeng 2024; 17:295-303. [PMID: 39372552 PMCID: PMC11450125 DOI: 10.1007/s12195-024-00814-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 08/20/2024] [Indexed: 10/08/2024] Open
Abstract
Purpose Accelerating wound healing is a main consideration in surgery. The three stages of wound healing are inflammatory response, tissue repair and cell proliferation. Much research has focused on epidermal cell proliferation and migration because this is an essential step in wound healing. Methods and Results The current study discovered that exosomes from Adipose-derived stem cell (ADSC) following hypoxic preconditioning (HExo) have a greater promotional effect on vaginal wound healing. Protein kinase B (AKT)/hypoxia-inducible factor 1-alpha (HIF-1α) play an important role in HExo-mediated HaCaT cell migration and proliferation. The promotional effect of HExo on rat wound healing was reversed by both, HIF‑1α and AKT inhibition. Phosphorylation of AKT (p-AKT) or HIF‑1α suppression reversed the protective effect of HExo on vaginal wound healing. Conclusion Taken together, our study found that hypoxic preconditioning of adipose MSC exosomes enhances vaginal wound healing via accelerated keratinocyte proliferation and migration through AKT/HIF‑1α axis activation.
Collapse
Affiliation(s)
- Xiaoyun Yang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji University School of Medicine, No. 389 Xincun Road, Shanghai, 200065 People’s Republic of China
| | - Shasha Zhang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji University School of Medicine, No. 389 Xincun Road, Shanghai, 200065 People’s Republic of China
| | - Kewei Chen
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji University School of Medicine, No. 389 Xincun Road, Shanghai, 200065 People’s Republic of China
| | - Dongsheng Shen
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji University School of Medicine, No. 389 Xincun Road, Shanghai, 200065 People’s Republic of China
| | - Yang Yang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji University School of Medicine, No. 389 Xincun Road, Shanghai, 200065 People’s Republic of China
| | - Aiqun Shen
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji University School of Medicine, No. 389 Xincun Road, Shanghai, 200065 People’s Republic of China
| | - Junhua Liang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji University School of Medicine, No. 389 Xincun Road, Shanghai, 200065 People’s Republic of China
| | - Mengjiao Xu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji University School of Medicine, No. 389 Xincun Road, Shanghai, 200065 People’s Republic of China
| | - Yuanyuan Yang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji University School of Medicine, No. 389 Xincun Road, Shanghai, 200065 People’s Republic of China
| | - Yanhong Zhao
- Department of Anesthesiology, Tongji Hospital, Tongji University School of Medicine, No. 389 Xincun Road, Shanghai, 200065 People’s Republic of China
| | - Huaifang Li
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji University School of Medicine, No. 389 Xincun Road, Shanghai, 200065 People’s Republic of China
| | - Xiaowen Tong
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji University School of Medicine, No. 389 Xincun Road, Shanghai, 200065 People’s Republic of China
| |
Collapse
|
4
|
Kemaloğlu CA, Dursun EN, Yay AH, Gökdemir NS, Mat ÖC, Gönen ZB. The Optimal Effective Dose of Adipose-Derived Stem Cell Exosomes in Wound Healing. Ann Plast Surg 2024; 93:253-260. [PMID: 39023411 DOI: 10.1097/sap.0000000000004032] [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: 07/20/2024]
Abstract
INTRODUCTION Although the effect of adipose-derived mesenchymal stem cell exosomes (ADSC-exos) on wound healing with different doses are shown in various studies, efficient and sufficient doses of ADSC-exos are still unknown. The study aimed to determine the optimal dose of ADSC-exos in wound healing. METHODS The 45 Sprague-Dawley rats were randomly divided into five groups, with seven animals in each. After dorsal circular defects were created, each wound was injected as follows: group 1: saline, group 2: 10 μg/mL of ADSC-exos, group 3: 100 μg/mL of ADSC-exos, group 4: 200 μg/mL of ADSC-exos, and group 5: 400 μg/mL of ADSC-exos. The effects of ADSC-exos on epithelization, angiogenesis, and collagen formation were analyzed macroscopically, histopathologically, and immunohistochemically on day 14. RESULTS A total of 200 μg/mL and 400 μg/mL ADSC-exos groups had higher epithelial tongue length, epithelial tongue area, and angiogenesis scores than the other groups. Although there was no statistical difference in fibrosis scores among groups, collagen fibers were becoming well-organized as the ADSC-exos doses increased. While the wound area was clinically smaller in the 200 μg/mL ADSC-exos group, there was no statistically significant difference among groups on day 14. CONCLUSIONS A total of 200 μg/mL of ADSC-exos was found to be the adequate and effective dose for re-epithelialization and angiogenesis in cutaneous wound healing. Moreover, the collagen density increased with a more regular pattern in the 200 μg/mL group, which can be important in scar regulation.
Collapse
Affiliation(s)
- Cemal Alper Kemaloğlu
- From the Department of Plastic, Reconstructive and Aesthetic Surgery, Erciyes University, Kayseri, Turkiye
| | - Ece Nur Dursun
- Department of Plastic, Reconstructive and Aesthetic Surgery, Kayseri Training and Research Hospital, Kayseri, Turkiye
| | - Arzu Hanım Yay
- Department of Histology and Embryology, Erciyes University, Kayseri, Turkiye
| | - Nur Seda Gökdemir
- Genome and Stem Cell Centre (GENKOK), Erciyes University, Kayseri, Turkiye
| | - Özge Cengiz Mat
- Department of Histology and Embryology, Erciyes University, Kayseri, Turkiye
| | - Zeynep Burçin Gönen
- Department of Oral and Maxillofacial Surgery, Genome and Stem Cell Centre, Erciyes University, Kayseri, Turkiye
| |
Collapse
|
5
|
Souza ILM, Suzukawa AA, Josino R, Marcon BH, Robert AW, Shigunov P, Correa A, Stimamiglio MA. Cellular In Vitro Responses Induced by Human Mesenchymal Stem/Stromal Cell-Derived Extracellular Vesicles Obtained from Suspension Culture. Int J Mol Sci 2024; 25:7605. [PMID: 39062847 PMCID: PMC11277484 DOI: 10.3390/ijms25147605] [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: 06/07/2024] [Revised: 07/04/2024] [Accepted: 07/09/2024] [Indexed: 07/28/2024] Open
Abstract
Mesenchymal stem/stromal cells (MSCs) and their extracellular vesicles (MSC-EVs) have been described to have important roles in tissue regeneration, including tissue repair, control of inflammation, enhancing angiogenesis, and regulating extracellular matrix remodeling. MSC-EVs have many advantages for use in regeneration therapies such as facility for dosage, histocompatibility, and low immunogenicity, thus possessing a lower possibility of rejection. In this work, we address the potential activity of MSC-EVs isolated from adipose-derived MSCs (ADMSC-EVs) cultured on cross-linked dextran microcarriers, applied to test the scalability and reproducibility of EV production. Isolated ADMSC-EVs were added into cultured human dermal fibroblasts (NHDF-1), keratinocytes (HaCat), endothelial cells (HUVEC), and THP-1 cell-derived macrophages to evaluate cellular responses (i.e., cell proliferation, cell migration, angiogenesis induction, and macrophage phenotype-switching). ADMSC viability and phenotype were assessed during cell culture and isolated ADMSC-EVs were monitored by nanotracking particle analysis, electron microscopy, and immunophenotyping. We observed an enhancement of HaCat proliferation; NHDF-1 and HaCat migration; endothelial tube formation on HUVEC; and the expression of inflammatory cytokines in THP-1-derived macrophages. The increased expression of TGF-β and IL-1β was observed in M1 macrophages treated with higher doses of ADMSC-EVs. Hence, EVs from microcarrier-cultivated ADMSCs are shown to modulate cell behavior, being able to induce skin tissue related cells to migrate and proliferate as well as stimulate angiogenesis and cause balance between pro- and anti-inflammatory responses in macrophages. Based on these findings, we suggest that the isolation of EVs from ADMSC suspension cultures makes it possible to induce in vitro cellular responses of interest and obtain sufficient particle numbers for the development of in vivo concept tests for tissue regeneration studies.
Collapse
Affiliation(s)
- Ingrid L. M. Souza
- Laboratory of Basic Biology of Stem Cells (Labcet), Carlos Chagas Institute, Fiocruz, Curitiba 81350-010, PR, Brazil (A.A.S.); (B.H.M.); (A.W.R.); (P.S.)
| | - Andreia A. Suzukawa
- Laboratory of Basic Biology of Stem Cells (Labcet), Carlos Chagas Institute, Fiocruz, Curitiba 81350-010, PR, Brazil (A.A.S.); (B.H.M.); (A.W.R.); (P.S.)
| | - Raphaella Josino
- Albert Einstein Israelite Hospital, São Paulo 05652-900, SP, Brazil
| | - Bruna H. Marcon
- Laboratory of Basic Biology of Stem Cells (Labcet), Carlos Chagas Institute, Fiocruz, Curitiba 81350-010, PR, Brazil (A.A.S.); (B.H.M.); (A.W.R.); (P.S.)
- Confocal and Electronic Microscopy Facility (RPT07C), Carlos Chagas Institute, Fiocruz, Curitiba 81350-010, PR, Brazil
| | - Anny W. Robert
- Laboratory of Basic Biology of Stem Cells (Labcet), Carlos Chagas Institute, Fiocruz, Curitiba 81350-010, PR, Brazil (A.A.S.); (B.H.M.); (A.W.R.); (P.S.)
- Confocal and Electronic Microscopy Facility (RPT07C), Carlos Chagas Institute, Fiocruz, Curitiba 81350-010, PR, Brazil
| | - Patrícia Shigunov
- Laboratory of Basic Biology of Stem Cells (Labcet), Carlos Chagas Institute, Fiocruz, Curitiba 81350-010, PR, Brazil (A.A.S.); (B.H.M.); (A.W.R.); (P.S.)
| | - Alejandro Correa
- Laboratory of Basic Biology of Stem Cells (Labcet), Carlos Chagas Institute, Fiocruz, Curitiba 81350-010, PR, Brazil (A.A.S.); (B.H.M.); (A.W.R.); (P.S.)
| | - Marco A. Stimamiglio
- Laboratory of Basic Biology of Stem Cells (Labcet), Carlos Chagas Institute, Fiocruz, Curitiba 81350-010, PR, Brazil (A.A.S.); (B.H.M.); (A.W.R.); (P.S.)
| |
Collapse
|
6
|
Qian L, Li B, Pi L, Fang B, Meng X. Hypoxic adipose stem cell-derived exosomes carrying high-abundant USP22 facilitate cutaneous wound healing through stabilizing HIF-1α and upregulating lncRNA H19. FASEB J 2024; 38:e23653. [PMID: 38738548 DOI: 10.1096/fj.202301403rr] [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: 07/11/2023] [Revised: 04/10/2024] [Accepted: 04/22/2024] [Indexed: 05/14/2024]
Abstract
Hypoxic preconditioning has been recognized as a promotive factor for accelerating cutaneous wound healing. Our previous study uncovered that exosomal lncRNA H19, derived from adipose-derived stem cells (ADSCs), plays a crucial role in orchestrating cutaneous wound healing. Herein, we aimed to explore whether there is a connection between hypoxia and ADSC-derived exosomes (ADSCs-exos) in cutaneous wound healing. Exosomes extracted from ADSCs under normoxic and hypoxic conditions were identified using transmission electron microscope (TEM) and particle size analysis. The effects of ADSCs-exos on the proliferation, migration, and angiogenesis of human umbilical vein endothelial cells (HUVECs) were evaluated by CCK-8, EdU, wound healing, and tube formation assays. Expression patterns of H19, HIF-1α, and USP22 were measured. Co-immunoprecipitation, chromatin immunoprecipitation, ubiquitination, and luciferase reporter assays were conducted to confirm the USP22/HIF-1α/H19 axis, which was further validated in a mice model of skin wound. Exosomes extracted from hypoxia-treated ADSCs (termed as H-ADSCs-exos) significantly increased cell proliferation, migration, and angiogenesis in H2O2-exposed HUVECs, and promoted cutaneous wound healing in vivo. Moreover, H-ADSCs and H-ADSCs-exos, which exhibited higher levels of H19, were found to be transcriptionally activated by HIF-1α. Mechanically, H-ADSCs carrying USP22 accounted for deubiquitinating and stabilizing HIF-1α. Additionally, H-ADSCs-exos improved cell proliferation, migration, and angiogenesis in H2O2-triggered HUVECs by activating USP22/HIF-1α axis and promoting H19 expression, which may provide a new clue for the clinical treatment of cutaneous wound healing.
Collapse
Affiliation(s)
- Li Qian
- Department of Plastic and Aesthetic (Burn) Surgery, the Second Xiangya Hospital, Central South University, Changsha, Hunan, P.R. China
| | - Bo Li
- Department of Plastic & Laser Cosmetic, Hunan Provincial People's Hospital, 1st Affiliated Hospital of Hunan Normal University, Changsha, Hunan, P.R. China
| | - Li Pi
- Department of Plastic and Aesthetic (Burn) Surgery, the Second Xiangya Hospital, Central South University, Changsha, Hunan, P.R. China
| | - Bairong Fang
- Department of Plastic and Aesthetic (Burn) Surgery, the Second Xiangya Hospital, Central South University, Changsha, Hunan, P.R. China
| | - Xianxi Meng
- Department of Plastic and Aesthetic (Burn) Surgery, the Second Xiangya Hospital, Central South University, Changsha, Hunan, P.R. China
| |
Collapse
|
7
|
Chi PL, Cheng CC, Wang MT, Liao JB, Kuo SH, Lin KC, Shen MC, Huang WC. Induced pluripotent stem cell-derived exosomes attenuate vascular remodelling in pulmonary arterial hypertension by targeting HIF-1α and Runx2. Cardiovasc Res 2024; 120:203-214. [PMID: 38252891 DOI: 10.1093/cvr/cvad185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 07/19/2023] [Accepted: 08/11/2023] [Indexed: 01/24/2024] Open
Abstract
AIMS Pulmonary arterial hypertension (PAH) is characterized by extensive pulmonary arterial remodelling. Although mesenchymal stem cell (MSC)-derived exosomes provide protective effects in PAH, MSCs exhibit limited senescence during in vitro expansion compared with the induced pluripotent stem cells (iPSCs). Moreover, the exact mechanism is not known. METHODS AND RESULTS In this study, we used murine iPSCs generated from mouse embryonic fibroblasts with triple factor (Oct4, Klf4, and Sox2) transduction to determine the efficacy and action mechanism of iPSC-derived exosomes (iPSC-Exo) in attenuating PAH in rats with monocrotaline (MCT)-induced pulmonary hypertension. Both early and late iPSC-Exo treatment effectively prevented the wall thickening and muscularization of pulmonary arterioles, improved the right ventricular systolic pressure, and alleviated the right ventricular hypertrophy in MCT-induced PAH rats. Pulmonary artery smooth muscle cells (PASMC) derived from MCT-treated rats (MCT-PASMC) developed more proliferative and pro-migratory phenotypes, which were attenuated by the iPSC-Exo treatment. Moreover, the proliferation and migration of MCT-PASMC were reduced by iPSC-Exo with suppression of PCNA, cyclin D1, MMP-1, and MMP-10, which are mediated via the HIF-1α and P21-activated kinase 1/AKT/Runx2 pathways. CONCLUSION IPSC-Exo are effective at reversing pulmonary hypertension by reducing pulmonary vascular remodelling and may provide an iPSC-free therapy for the treatment of PAH.
Collapse
Affiliation(s)
- Pei-Ling Chi
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
- Department of Pathology and Laboratory Medicine, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
| | - Chin-Chang Cheng
- Department of Internal Medicine, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
| | - Mei-Tzu Wang
- Department of Critical Care Medicine, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
| | - Jia-Bin Liao
- Department of Pathology and Laboratory Medicine, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
| | - Shu-Hung Kuo
- Department of Critical Care Medicine, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
| | - Kun-Chang Lin
- Department of Critical Care Medicine, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
| | - Min-Ci Shen
- Department of Critical Care Medicine, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
| | - Wei-Chun Huang
- Department of Critical Care Medicine, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
- Department of Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Department of Physical Therapy, Fooyin University, Kaohsiung, Taiwan
| |
Collapse
|
8
|
Miron RJ, Estrin NE, Sculean A, Zhang Y. Understanding exosomes: Part 2-Emerging leaders in regenerative medicine. Periodontol 2000 2024; 94:257-414. [PMID: 38591622 DOI: 10.1111/prd.12561] [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: 02/04/2024] [Revised: 02/16/2024] [Accepted: 02/21/2024] [Indexed: 04/10/2024]
Abstract
Exosomes are the smallest subset of extracellular signaling vesicles secreted by most cells with the ability to communicate with other tissues and cell types over long distances. Their use in regenerative medicine has gained tremendous momentum recently due to their ability to be utilized as therapeutic options for a wide array of diseases/conditions. Over 5000 publications are currently being published yearly on this topic, and this number is only expected to dramatically increase as novel therapeutic strategies continue to be developed. Today exosomes have been applied in numerous contexts including neurodegenerative disorders (Alzheimer's disease, central nervous system, depression, multiple sclerosis, Parkinson's disease, post-traumatic stress disorders, traumatic brain injury, peripheral nerve injury), damaged organs (heart, kidney, liver, stroke, myocardial infarctions, myocardial infarctions, ovaries), degenerative processes (atherosclerosis, diabetes, hematology disorders, musculoskeletal degeneration, osteoradionecrosis, respiratory disease), infectious diseases (COVID-19, hepatitis), regenerative procedures (antiaging, bone regeneration, cartilage/joint regeneration, osteoarthritis, cutaneous wounds, dental regeneration, dermatology/skin regeneration, erectile dysfunction, hair regrowth, intervertebral disc repair, spinal cord injury, vascular regeneration), and cancer therapy (breast, colorectal, gastric cancer and osteosarcomas), immune function (allergy, autoimmune disorders, immune regulation, inflammatory diseases, lupus, rheumatoid arthritis). This scoping review is a first of its kind aimed at summarizing the extensive regenerative potential of exosomes over a broad range of diseases and disorders.
Collapse
Affiliation(s)
- Richard J Miron
- Department of Periodontology, University of Bern, Bern, Switzerland
| | - Nathan E Estrin
- Advanced PRF Education, Venice, Florida, USA
- School of Dental Medicine, Lake Erie College of Osteopathic Medicine, Bradenton, Florida, USA
| | - Anton Sculean
- Department of Periodontology, University of Bern, Bern, Switzerland
| | - Yufeng Zhang
- Department of Oral Implantology, University of Wuhan, Wuhan, China
| |
Collapse
|
9
|
Tan F, Li X, Wang Z, Li J, Shahzad K, Zheng J. Clinical applications of stem cell-derived exosomes. Signal Transduct Target Ther 2024; 9:17. [PMID: 38212307 PMCID: PMC10784577 DOI: 10.1038/s41392-023-01704-0] [Citation(s) in RCA: 70] [Impact Index Per Article: 70.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Revised: 10/15/2023] [Accepted: 11/12/2023] [Indexed: 01/13/2024] Open
Abstract
Although stem cell-based therapy has demonstrated considerable potential to manage certain diseases more successfully than conventional surgery, it nevertheless comes with inescapable drawbacks that might limit its clinical translation. Compared to stem cells, stem cell-derived exosomes possess numerous advantages, such as non-immunogenicity, non-infusion toxicity, easy access, effortless preservation, and freedom from tumorigenic potential and ethical issues. Exosomes can inherit similar therapeutic effects from their parental cells such as embryonic stem cells and adult stem cells through vertical delivery of their pluripotency or multipotency. After a thorough search and meticulous dissection of relevant literature from the last five years, we present this comprehensive, up-to-date, specialty-specific and disease-oriented review to highlight the surgical application and potential of stem cell-derived exosomes. Exosomes derived from stem cells (e.g., embryonic, induced pluripotent, hematopoietic, mesenchymal, neural, and endothelial stem cells) are capable of treating numerous diseases encountered in orthopedic surgery, neurosurgery, plastic surgery, general surgery, cardiothoracic surgery, urology, head and neck surgery, ophthalmology, and obstetrics and gynecology. The diverse therapeutic effects of stem cells-derived exosomes are a hierarchical translation through tissue-specific responses, and cell-specific molecular signaling pathways. In this review, we highlight stem cell-derived exosomes as a viable and potent alternative to stem cell-based therapy in managing various surgical conditions. We recommend that future research combines wisdoms from surgeons, nanomedicine practitioners, and stem cell researchers in this relevant and intriguing research area.
Collapse
Affiliation(s)
- Fei Tan
- Department of ORL-HNS, Shanghai Fourth People's Hospital, and School of Medicine, Tongji University, Shanghai, China.
- Plasma Medicine and Surgical Implants Center, Tongji University, Shanghai, China.
- The Royal College of Surgeons in Ireland, Dublin, Ireland.
- The Royal College of Surgeons of England, London, UK.
| | - Xuran Li
- Department of ORL-HNS, Shanghai Fourth People's Hospital, and School of Medicine, Tongji University, Shanghai, China
- Plasma Medicine and Surgical Implants Center, Tongji University, Shanghai, China
| | - Zhao Wang
- Department of ORL-HNS, Shanghai Fourth People's Hospital, and School of Medicine, Tongji University, Shanghai, China
| | - Jiaojiao Li
- Department of ORL-HNS, Shanghai Fourth People's Hospital, and School of Medicine, Tongji University, Shanghai, China
- Plasma Medicine and Surgical Implants Center, Tongji University, Shanghai, China
| | - Khawar Shahzad
- Department of ORL-HNS, Shanghai Fourth People's Hospital, and School of Medicine, Tongji University, Shanghai, China
- Plasma Medicine and Surgical Implants Center, Tongji University, Shanghai, China
| | - Jialin Zheng
- Center for Translational Neurodegeneration and Regenerative Therapy, Tongji Hospital affiliated to Tongji University School of Medicine, Shanghai, China
- Shanghai Frontiers Science Center of Nanocatalytic Medicine, Tongji University, Shanghai, China
| |
Collapse
|
10
|
Chang H, Chen J, Ding K, Cheng T, Tang S. Highly-expressed lncRNA FOXD2-AS1 in adipose mesenchymal stem cell derived exosomes affects HaCaT cells via regulating miR-185-5p/ROCK2 axis. Adipocyte 2023; 12:2173513. [PMID: 36775902 PMCID: PMC9928455 DOI: 10.1080/21623945.2023.2173513] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/14/2023] Open
Abstract
The healing of skin wounds is a highly coordinated multi-step process that occurs after trauma including surgical incisions, thermal burns, and chronic ulcers. In this study, the authors investigated lncRNA FOXD2-AS1 function in adipose mesenchymal exosomes from ADMSCs that were successfully extracted. Highly expressed lncRNA FOXD2-AS1 in ADMSCs-exosomes accelerated HaCaT cell migration and proliferation. LncRNA FOXD2-AS1 negatively targeted miR-185-5p, and miR-185-5p negatively targeted ROCK2. Highly expressed lncRNA FOXD2-AS1 in ADMSCs-exosomes promoted HaCaT cell migration and proliferation via down-regulating miR-185-5p and further up-regulating ROCK2. In conclusion, LncRNA FOXD2-AS1 overexpression in ADMSCs derived exosomes might accelerate HaCaT cell migration and proliferation via modulating the miR-185-5p/ROCK2 axis.
Collapse
Affiliation(s)
- Huanchao Chang
- Plastic Surgery of Plastic Surgery Hospital, Weifang Medical University, Weifang, China
| | - Junliang Chen
- Vascular surgery department, Affiliated Hospital of Weifang Medical College, Weifang, China
| | - Kun Ding
- Plastic Surgery of Plastic Surgery Hospital, Weifang Medical University, Weifang, China
| | - Tianling Cheng
- Burn plastic surgery, The First Affiliated Hospital of Xi’an Medical University, Xi’an, China
| | - Shengjian Tang
- Plastic Surgery of Plastic Surgery Hospital, Weifang Medical University, Weifang, China,CONTACT Shengjian Tang Plastic Surgery Institute, Weifang Medical University, 4948 Shengli East Street, Kuiwen District, Weifang, 261041, China
| |
Collapse
|
11
|
Wen W, Yang L, Wang X, Zhang H, Wu F, Xu K, Chen S, Liao Z. Fucoidan promotes angiogenesis and accelerates wound healing through AKT/Nrf2/HIF-1α signalling pathway. Int Wound J 2023; 20:3606-3618. [PMID: 37203309 PMCID: PMC10588368 DOI: 10.1111/iwj.14239] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 05/07/2023] [Accepted: 05/09/2023] [Indexed: 05/20/2023] Open
Abstract
After skin injury, wound repair involves a complex process in which angiogenesis plays a crucial role. Previous research has indicated that fucoidan may aid in wound healing; we therefore hypothesised that fucoidan may speed up the process by promoting angiogenesis. In this study, we investigated the potential molecular mechanism underlying fucoidan's ability to accelerate wound healing by promoting angiogenesis. Using a full-cut wound model, we observed that fucoidan significantly intensified wound closure and promoted granulation formation and collagen deposition. Immunofluorescence staining revealed that fucoidan also promoted wound angiogenesis, specifically by accelerating the migration of new blood vessels to the middle area of the wound. Furthermore, fucoidan demonstrated the ability to enhance the proliferation of human umbilical vein endothelial cells (HUVECs) damaged by hydrogen peroxide (H2 O2 ) and to improve the formation of endothelial tubes. Mechanistic studies revealed that fucoidan upregulated the protein levels of the AKT/Nrf2/HIF-1α signalling pathway, which plays a crucial role in angiogenesis. This was further confirmed using the inhibitor LY294002, which reversed the promotion of endothelial tube formation by fucoidan. Overall, our findings suggest that fucoidan can promote angiogenesis via the AKT/Nrf2/HIF-1α signalling pathway and accelerate wound healing.
Collapse
Affiliation(s)
- Wenting Wen
- College of Life and Environmental SciencesWenzhou UniversityZhejiangChina
| | - Liangliang Yang
- School of Pharmaceutical Sciences, Wenzhou Wound Repair and Regeneration Key Laboratory, Cixi Biomedical Research InstituteWenzhou Medical UniversityZhejiangChina
| | - Xin Wang
- Dpartment of Plastic and Reconstructive Surgery, Hand and MicrosurgeryNingbo NO.6 HospitalZhejiangChina
| | - Hongyu Zhang
- School of Pharmaceutical Sciences, Wenzhou Wound Repair and Regeneration Key Laboratory, Cixi Biomedical Research InstituteWenzhou Medical UniversityZhejiangChina
| | - Fangfang Wu
- Department of Emergency, The Second Affiliated Hospital and Yuying Children's HospitalWenzhou Medical UniversityWenzhouChina
| | - Ke Xu
- College of Life and Environmental SciencesWenzhou UniversityZhejiangChina
| | - Shaodong Chen
- Department of OrthopaedicsLishui People's HospitalZhejiangChina
| | - Zhiyong Liao
- College of Life and Environmental SciencesWenzhou UniversityZhejiangChina
| |
Collapse
|
12
|
Ni H, Xi J, Tang J, Yan Y, Chu Y, Zhou J. Therapeutic Potential of Extracellular Vesicles from Different Stem Cells in Chronic Wound Healing. Stem Cell Rev Rep 2023; 19:1596-1614. [PMID: 37178227 DOI: 10.1007/s12015-023-10540-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/31/2023] [Indexed: 05/15/2023]
Abstract
Wound healing has long been a complex problem, especially in chronic wounds. Although debridement, skin grafting, and antimicrobial dressings have been used to treat chronic wounds, their treatment period is long, expensive, and has specific rejection reactions. The poor treatment results of traditional methods have caused psychological stress to patients and a substantial economic burden to society. Extracellular vesicles (EVs) are nanoscale vesicles secreted by cells. They play an essential role in intercellular communication. Numerous studies have confirmed that stem cell-derived extracellular vesicles (SC-EVs) can inhibit overactive inflammation, induce angiogenesis, promote re-epithelization, and reduce scar formation. Therefore, SC-EVs are expected to be a novel cell-free strategy for chronic wound treatment. We first summarize the pathological factors that hinder wound healing and discuss how SC-EVs accelerate chronic wound repair. And then, we also compare the advantages and disadvantages of different SC-EVs for chronic wound treatment. Finally, we discuss the limitations of SC-EVs usage and provide new thoughts for future SC-EVs research in chronic wound treatment.
Collapse
Affiliation(s)
- Haoxi Ni
- School of Medicine, Jiangsu University, Zhenjiang, 212013, China
| | - Jianbo Xi
- Wujin Institute of Molecular Diagnostics and Precision Cancer Medicine of Jiangsu University, Changzhou, 213017, China
- Changzhou Key Laboratory of Molecular Diagnostics and Precision Cancer Medicine, Changzhou, 213017, China
| | - Jianjun Tang
- Wujin Institute of Molecular Diagnostics and Precision Cancer Medicine of Jiangsu University, Changzhou, 213017, China
- Changzhou Key Laboratory of Molecular Diagnostics and Precision Cancer Medicine, Changzhou, 213017, China
- Department of General Surgery, Wujin Clinical College of Xuzhou Medical University, Changzhou, 213017, China
| | - Yongmin Yan
- Wujin Institute of Molecular Diagnostics and Precision Cancer Medicine of Jiangsu University, Changzhou, 213017, China
- Changzhou Key Laboratory of Molecular Diagnostics and Precision Cancer Medicine, Changzhou, 213017, China
- Department of Laboratory Medicine, Wujin Hospital Affiliated with Jiangsu University, Changzhou, 213017, China
| | - Ying Chu
- Wujin Institute of Molecular Diagnostics and Precision Cancer Medicine of Jiangsu University, Changzhou, 213017, China.
- Changzhou Key Laboratory of Molecular Diagnostics and Precision Cancer Medicine, Changzhou, 213017, China.
| | - Jing Zhou
- Wujin Institute of Molecular Diagnostics and Precision Cancer Medicine of Jiangsu University, Changzhou, 213017, China.
- Changzhou Key Laboratory of Molecular Diagnostics and Precision Cancer Medicine, Changzhou, 213017, China.
| |
Collapse
|
13
|
Sousa P, Lopes B, Sousa AC, Moreira A, Coelho A, Alvites R, Alves N, Geuna S, Maurício AC. Advancements and Insights in Exosome-Based Therapies for Wound Healing: A Comprehensive Systematic Review (2018-June 2023). Biomedicines 2023; 11:2099. [PMID: 37626596 PMCID: PMC10452374 DOI: 10.3390/biomedicines11082099] [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: 06/25/2023] [Revised: 07/14/2023] [Accepted: 07/22/2023] [Indexed: 08/27/2023] Open
Abstract
Exosomes have shown promising potential as a therapeutic approach for wound healing. Nevertheless, the translation from experimental studies to commercially available treatments is still lacking. To assess the current state of research in this field, a systematic review was performed involving studies conducted and published over the past five years. A PubMed search was performed for English-language, full-text available papers published from 2018 to June 2023, focusing on exosomes derived from mammalian sources and their application in wound healing, particularly those involving in vivo assays. Out of 531 results, 148 papers were selected for analysis. The findings revealed that exosome-based treatments improve wound healing by increasing angiogenesis, reepithelization, collagen deposition, and decreasing scar formation. Furthermore, there was significant variability in terms of cell sources and types, biomaterials, and administration routes under investigation, indicating the need for further research in this field. Additionally, a comparative examination encompassing diverse cellular origins, types, administration pathways, or biomaterials is imperative. Furthermore, the predominance of rodent-based animal models raises concerns, as there have been limited advancements towards more complex in vivo models and scale-up assays. These constraints underscore the substantial efforts that remain necessary before attaining commercially viable and extensively applicable therapeutic approaches using exosomes.
Collapse
Affiliation(s)
- Patrícia Sousa
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, No. 228, 4050-313 Porto, Portugal; (P.S.); (B.L.); (A.C.S.); (A.M.); (A.C.); (R.A.)
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado 55142, 4051-401 Porto, Portugal
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), 1300-477 Lisboa, Portugal
| | - Bruna Lopes
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, No. 228, 4050-313 Porto, Portugal; (P.S.); (B.L.); (A.C.S.); (A.M.); (A.C.); (R.A.)
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado 55142, 4051-401 Porto, Portugal
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), 1300-477 Lisboa, Portugal
| | - Ana Catarina Sousa
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, No. 228, 4050-313 Porto, Portugal; (P.S.); (B.L.); (A.C.S.); (A.M.); (A.C.); (R.A.)
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado 55142, 4051-401 Porto, Portugal
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), 1300-477 Lisboa, Portugal
| | - Alícia Moreira
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, No. 228, 4050-313 Porto, Portugal; (P.S.); (B.L.); (A.C.S.); (A.M.); (A.C.); (R.A.)
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado 55142, 4051-401 Porto, Portugal
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), 1300-477 Lisboa, Portugal
| | - André Coelho
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, No. 228, 4050-313 Porto, Portugal; (P.S.); (B.L.); (A.C.S.); (A.M.); (A.C.); (R.A.)
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado 55142, 4051-401 Porto, Portugal
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), 1300-477 Lisboa, Portugal
| | - Rui Alvites
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, No. 228, 4050-313 Porto, Portugal; (P.S.); (B.L.); (A.C.S.); (A.M.); (A.C.); (R.A.)
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado 55142, 4051-401 Porto, Portugal
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), 1300-477 Lisboa, Portugal
- Instituto Universitário de Ciências da Saúde (CESPU), Avenida Central de Gandra 1317, 4585-116 Paredes, Portugal
| | - Nuno Alves
- Centre for Rapid and Sustainable Product Development, Polytechnic of Leiria, 2430-028 Marinha Grande, Portugal;
| | - Stefano Geuna
- Department of Clinical and Biological Sciences, Cavalieri Ottolenghi Neuroscience Institute, University of Turin, Ospedale San Luigi, 10043 Turin, Italy;
| | - Ana Colette Maurício
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, No. 228, 4050-313 Porto, Portugal; (P.S.); (B.L.); (A.C.S.); (A.M.); (A.C.); (R.A.)
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado 55142, 4051-401 Porto, Portugal
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), 1300-477 Lisboa, Portugal
| |
Collapse
|
14
|
Zhou C, Zhang B, Yang Y, Jiang Q, Li T, Gong J, Tang H, Zhang Q. Stem cell-derived exosomes: emerging therapeutic opportunities for wound healing. Stem Cell Res Ther 2023; 14:107. [PMID: 37101197 PMCID: PMC10134577 DOI: 10.1186/s13287-023-03345-0] [Citation(s) in RCA: 58] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 04/14/2023] [Indexed: 04/28/2023] Open
Abstract
Wound healing is a dynamic and highly sequential process involving a series of overlapping spatial and temporal phases, including hemostasis, inflammation, proliferation, and tissue remodeling. Mesenchymal stem cells (MSCs) are multipotent stem cells with self-renewal, multidirectional differentiation potential, and paracrine regulation. Exosomes are subcellular vesicular components 30-150 nm in size and are novel carriers of intercellular communication in regulating the biological behaviors of skin cells. Compared to MSCs, MSC-derived exosomes (MSC-exos) possess lower immunogenicity, easy storage, and highly effective biological activity. MSC-exos, mainly derived from adipose-derived stem cells (ADSCs), bone marrow-derived MSCs (BMSCs), human umbilical cord MSCs (hUC-MSCs), and other stem cell types, play a role in shaping the activity of fibroblasts, keratinocytes, immune cells, and endothelial cells in diabetic wounds, inflammatory wound repair, and even wound-related keloid formation. Therefore, this study focuses on the specific roles and mechanisms of different MSC-exos in wound healing, as well as the current limitations and various perspectives. Deciphering the biological properties of MSC-exos is crucial to providing a promising cell-free therapeutic tool for wound healing and cutaneous regeneration.
Collapse
Affiliation(s)
- Chuchao Zhou
- Department of Plastic Surgery, Wuhan Third Hospital (Tongren Hospital of Wuhan University), Wuhan, 430060, China
| | - Boyu Zhang
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, Hubei, China
| | - Yanqing Yang
- Department of Plastic Surgery, Wuhan Third Hospital (Tongren Hospital of Wuhan University), Wuhan, 430060, China
| | - Qiong Jiang
- Department of Pharmacy, Xianning Central Hospital, The First Affiliated Hospital of Hubei University of Science and Technology, Xianning, 437000, Hubei, China
| | - Tianyu Li
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jun Gong
- Department of Biliary-Pancreatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, Hubei, China.
| | - Hongbo Tang
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, Hubei, China.
| | - Qi Zhang
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, Hubei, China.
| |
Collapse
|
15
|
Adipose-Derived Stem Cell Extracellular Vesicles Improve Wound Closure and Angiogenesis in Diabetic Mice. Plast Reconstr Surg 2023; 151:331-342. [PMID: 36696316 DOI: 10.1097/prs.0000000000009840] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
BACKGROUND Currently, there is a lack in therapy that promotes the reepithelialization of diabetic wounds as an alternative to skin grafting. Here, the authors hypothesized that extracellular vesicles from adipose-derived stem cells (ADSC-EVs) could accelerate wound closure through rescuing the function of keratinocytes in diabetic mice. METHODS The effect of ADSC-EVs on the biological function of human keratinocyte cells was assayed in vitro. In vivo, 81 male severe combined immune deficiency mice aged 8 weeks were divided randomly into the extracellular vesicle-treated diabetes group (n = 27), the phosphate-buffered saline-treated diabetes group (n = 27), and the phosphate-buffered saline-treated normal group (n = 27). A round, 8-mm-diameter, full-skin defect was performed on the back skin of each mouse. The wound closure kinetics, average healing time, reepithelialization rate, and neovascularization were evaluated by histological staining. RESULTS In vitro, ADSC-EVs improved proliferation, migration, and proangiogenic potential, and inhibited the apoptosis of human keratinocyte cells by suppressing Fasl expression with the optimal dose of 40 μg/mL. In vivo, postoperative dripping of ADSC-EVs at the dose of 40 μg/mL accelerated diabetic wound healing, with a 15.8% increase in closure rate and a 3.3-day decrease in average healing time. ADSC-EVs improved reepithelialization (18.2%) with enhanced epithelial proliferation and filaggrin expression, and suppressed epithelial apoptosis and Fasl expression. A 2.7-fold increase in the number of CD31-positive cells was also observed. CONCLUSION ADSC-EVs improve diabetic wound closure and angiogenesis by enhancing keratinocyte-mediated reepithelialization and vascularization. CLINICAL RELEVANCE STATEMENT ADSC-EVs could be developed as a regenerative medicine for diabetic wound care.
Collapse
|
16
|
Wu J, Chen LH, Sun SY, Li Y, Ran XW. Mesenchymal stem cell-derived exosomes: The dawn of diabetic wound healing. World J Diabetes 2022; 13:1066-1095. [PMID: 36578867 PMCID: PMC9791572 DOI: 10.4239/wjd.v13.i12.1066] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 11/04/2022] [Accepted: 11/23/2022] [Indexed: 12/15/2022] Open
Abstract
Chronic wound healing has long been an unmet medical need in the field of wound repair, with diabetes being one of the major etiologies. Diabetic chronic wounds (DCWs), especially diabetic foot ulcers, are one of the most threatening chronic complications of diabetes. Although the treatment strategies, drugs, and dressings for DCWs have made great progress, they remain ineffective in some patients with refractory wounds. Stem cell-based therapies have achieved specific efficacy in various fields, with mesenchymal stem cells (MSCs) being the most widely used. Although MSCs have achieved good feedback in preclinical studies and clinical trials in the treatment of cutaneous wounds or other situations, the potential safety concerns associated with allogeneic/autologous stem cells and unknown long-term health effects need further attention and supervision. Recent studies have reported that stem cells mainly exert their trauma repair effects through paracrine secretion, and exosomes play an important role in intercellular communication as their main bioactive component. MSC-derived exosomes (MSC-Exos) inherit the powerful inflammation and immune modulation, angiogenesis, cell proliferation and migration promotion, oxidative stress alleviation, collagen remodeling imbalances regulation of their parental cells, and can avoid the potential risks of direct stem cell transplantation to a large extent, thus demonstrating promising performance as novel "cell-free" therapies in chronic wounds. This review aimed to elucidate the potential mechanism and update the progress of MSC-Exos in DCW healing, thereby providing new therapeutic directions for DCWs that are difficult to be cured using conventional therapy.
Collapse
Affiliation(s)
- Jing Wu
- Innovation Center for Wound Repair, Diabetic Foot Care Center, Department of Endocrinology and Metabolism, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Li-Hong Chen
- Innovation Center for Wound Repair, Diabetic Foot Care Center, Department of Endocrinology and Metabolism, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Shi-Yi Sun
- Innovation Center for Wound Repair, Diabetic Foot Care Center, Department of Endocrinology and Metabolism, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Yan Li
- Innovation Center for Wound Repair, Diabetic Foot Care Center, Department of Endocrinology and Metabolism, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Xing-Wu Ran
- Innovation Center for Wound Repair, Diabetic Foot Care Center, Department of Endocrinology and Metabolism, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| |
Collapse
|
17
|
Subhan BS, Ki M, Verzella A, Shankar S, Rabbani PS. Behind the Scenes of Extracellular Vesicle Therapy for Skin Injuries and Disorders. Adv Wound Care (New Rochelle) 2022; 11:575-597. [PMID: 34806432 PMCID: PMC9419953 DOI: 10.1089/wound.2021.0066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 11/10/2021] [Indexed: 01/29/2023] Open
Abstract
Significance: Skin wounds and disorders compromise the protective functions of skin and patient quality of life. Although accessible on the surface, they are challenging to address due to paucity of effective therapies. Exogenous extracellular vesicles (EVs) and cell-free derivatives of adult multipotent stromal cells (MSCs) are developing as a treatment modality. Knowledge of origin MSCs, EV processing, and mode of action is necessary for directed use of EVs in preclinical studies and methodical translation. Recent Advances: Nanoscale to microscale EVs, although from nonskin cells, induce functional responses in cutaneous wound cellular milieu. EVs allow a shift from cell-based to cell-free/derived modalities by carrying the MSC beneficial factors but eliminating risks associated with MSC transplantation. EVs have demonstrated striking efficacy in resolution of preclinical wound models, specifically within the complexity of skin structure and wound pathology. Critical Issues: To facilitate comparison across studies, tissue sources and processing of MSCs, culture conditions, isolation and preparations of EVs, and vesicle sizes require standardization as these criteria influence EV types and contents, and potentially determine the induced biological responses. Procedural parameters for all steps preceding the actual therapeutic administration may be the key to generating EVs that demonstrate consistent efficacy through known mechanisms. We provide a comprehensive review of such parameters and the subsequent tissue, cellular and molecular impact of the derived EVs in different skin wounds/disorders. Future Directions: We will gain more complete knowledge of EV-induced effects in skin, and specificity for different wounds/conditions. The safety and efficacy of current preclinical xenogenic applications will favor translation into allogenic clinical applications of EVs as a biologic.
Collapse
Affiliation(s)
- Bibi S. Subhan
- Hansjörg Wyss Department of Plastic Surgery, New York University School of Medicine, New York, New York, USA
| | - Michelle Ki
- Hansjörg Wyss Department of Plastic Surgery, New York University School of Medicine, New York, New York, USA
| | - Alexandra Verzella
- Hansjörg Wyss Department of Plastic Surgery, New York University School of Medicine, New York, New York, USA
| | - Shruthi Shankar
- Hansjörg Wyss Department of Plastic Surgery, New York University School of Medicine, New York, New York, USA
| | - Piul S. Rabbani
- Hansjörg Wyss Department of Plastic Surgery, New York University School of Medicine, New York, New York, USA
| |
Collapse
|
18
|
Nallakumarasamy A, Jeyaraman M, Maffulli N, Jeyaraman N, Suresh V, Ravichandran S, Gupta M, Potty AG, El-Amin SF, Khanna M, Gupta A. Mesenchymal Stromal Cell-Derived Extracellular Vesicles in Wound Healing. Life (Basel) 2022; 12:1733. [PMID: 36362890 PMCID: PMC9699035 DOI: 10.3390/life12111733] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Accepted: 10/24/2022] [Indexed: 07/26/2023] Open
Abstract
The well-orchestrated process of wound healing may be negatively impacted from interrupted or incomplete tissue regenerative processes. The healing potential is further compromised in patients with diabetes mellitus, chronic venous insufficiency, critical limb ischemia, and immunocompromised conditions, with a high health care burden and expenditure. Stem cell-based therapy has shown promising results in clinical studies. Mesenchymal stem cell-derived exosomes (MSC Exos) may favorably impact intercellular signaling and immunomodulation, promoting neoangiogenesis, collagen synthesis, and neoepithelization. This article gives an outline of the biogenesis and mechanism of extracellular vesicles (EVs), particularly exosomes, in the process of tissue regeneration and discusses the use of preconditioned exosomes, platelet-rich plasma-derived exosomes, and engineered exosomes in three-dimensional bioscaffolds such as hydrogels (collagen and chitosan) to prolong the contact time of exosomes at the recipient site within the target tissue. An appropriate antibiotic therapy based on culture-specific guidance coupled with the knowledge of biopolymers helps to fabricate nanotherapeutic materials loaded with MSC Exos to effectively deliver drugs locally and promote novel approaches for the management of chronic wounds.
Collapse
Affiliation(s)
- Arulkumar Nallakumarasamy
- Department of Orthopaedics, All India Institute of Medical Sciences, Bhubaneswar 751019, Odissa, India
- Fellow in Orthopaedic Rheumatology, Dr. RML National Law University, Lucknow 226010, Uttar Pradesh, India
- Indian Stem Cell Study Group (ISCSG) Association, Lucknow 226010, Uttar Pradesh, India
| | - Madhan Jeyaraman
- Indian Stem Cell Study Group (ISCSG) Association, Lucknow 226010, Uttar Pradesh, India
- Department of Orthopaedics, Faculty of Medicine—Sri Lalithambigai Medical College and Hospital, Dr. MGR Educational and Research Institute, Chennai 600095, Tamil Nadu, India
- Department of Medical Research and Translational Medicine, Faculty of Medicine—Sri Lalithambigai Medical College and Hospital, Dr. MGR Educational and Research Institute, Chennai 600095, Tamil Nadu, India
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida 201310, Uttar Pradesh, India
- South Texas Orthopaedic Research Institute (STORI Inc.), Laredo, TX 78045, USA
| | - Nicola Maffulli
- Department of Musculoskeletal Disorders, School of Medicine and Surgery, University of Salerno, 84084 Fisciano, Italy
- San Giovanni di Dio e Ruggi D’Aragona Hospital “Clinica Ortopedica” Department, Hospital of Salerno, 84124 Salerno, Italy
- Barts and the London School of Medicine and Dentistry, Centre for Sports and Exercise Medicine, Queen Mary University of London, London E1 4DG, UK
- School of Pharmacy and Bioengineering, Keele University School of Medicine, Stoke on Trent ST5 5BG, UK
| | - Naveen Jeyaraman
- Indian Stem Cell Study Group (ISCSG) Association, Lucknow 226010, Uttar Pradesh, India
- Fellow in Joint Replacement, Department of Orthopaedics, Atlas Hospitals, Tiruchirappalli 620002, Tamil Nadu, India
| | - Veerasivabalan Suresh
- Department of Obstetrics-Gynecology, Madras Medical College and Hospital, Chennai 600003, Tamil Nadu, India
| | - Srinath Ravichandran
- Department of General and GI Surgery, Stepping Hill Hospital, Stockport NHS Foundation Trust, Stockport SK27JE, UK
| | - Manu Gupta
- Polar Aesthetics Dental & Cosmetic Centre, Noida 201301, Uttar Pradesh, India
| | - Anish G. Potty
- South Texas Orthopaedic Research Institute (STORI Inc.), Laredo, TX 78045, USA
| | - Saadiq F. El-Amin
- El-Amin Orthopaedic & Sports Medicine Institute, Lawrenceville, GA 30043, USA
- Regenerative Sports Medicine, Lawrenceville, GA 30043, USA
- BioIntegrate, Lawrenceville, GA 30043, USA
| | - Manish Khanna
- Indian Stem Cell Study Group (ISCSG) Association, Lucknow 226010, Uttar Pradesh, India
- Department of Orthopaedics, Autonomous State Medical College, Ayodhya 224135, Uttar Pradesh, India
| | - Ashim Gupta
- Indian Stem Cell Study Group (ISCSG) Association, Lucknow 226010, Uttar Pradesh, India
- South Texas Orthopaedic Research Institute (STORI Inc.), Laredo, TX 78045, USA
- BioIntegrate, Lawrenceville, GA 30043, USA
- Regenerative Orthopaedics, Noida 201301, Uttar Pradesh, India
- Future Biologics, Lawrenceville, GA 30043, USA
| |
Collapse
|
19
|
Wang Y, Zhu J, Chen J, Xu R, Groth T, Wan H, Zhou G. The Signaling Pathways Induced by Exosomes in Promoting Diabetic Wound Healing: A Mini-Review. Curr Issues Mol Biol 2022; 44:4960-4976. [PMID: 36286052 PMCID: PMC9600352 DOI: 10.3390/cimb44100337] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 10/08/2022] [Accepted: 10/13/2022] [Indexed: 11/16/2022] Open
Abstract
Impaired healing of diabetic wounds harms patients' quality of life and even leads to disability and death, which is an urgent issue to be solved clinically. Despite the great progress that has been achieved, it remains a worldwide challenge to develop effective therapeutic treatments for diabetic wounds. Recently, exosomes have attracted special attention because they can be involved in immune response, antigen presentation, cell migration, cell differentiation, tumor invasion and other processes. Meanwhile, exosomes have been proven to hold great potential in the treatment of diabetic wounds. Mechanistic studies of exosomes based on signaling pathways could not only help to uncover the mechanisms by which exosomes promote diabetic wound healing but could also provide a theoretical basis for the clinical application of exosomes. Herein, our mini-review aims to summarize the progress of research on the use of various exosomes derived from different cell types to promote diabetic wound healing, with a focus on the classical signaling pathways, including PI3K/Akt, Wnt, NF-κB, MAPK, Notch, Nrf2, HIF-1α/VEGF and TGF-β/Smad. The results show that exosomes could regulate these signaling pathways to down-regulate inflammation, reduce oxidative stress, increase angiogenesis, promote fibroblast proliferation, induce re-epithelization and inhibit scar formation, making exosomes attractive candidates for the treatment of diabetic wounds.
Collapse
Affiliation(s)
- Yanying Wang
- The Second Clinical Medical College, Zhejiang Chinese Medical University, 548 Binwen Road, Hangzhou 310053, China
| | - Jiayan Zhu
- College of Life Science, Zhejiang Chinese Medical University, 548 Binwen Road, Hangzhou 310053, China
| | - Jing Chen
- College of Life Science, Zhejiang Chinese Medical University, 548 Binwen Road, Hangzhou 310053, China
| | - Ruojiao Xu
- College of Life Science, Zhejiang Chinese Medical University, 548 Binwen Road, Hangzhou 310053, China
| | - Thomas Groth
- Department Biomedical Materials, Institute of Pharmacy, Martin Luther University Halle-Wittenberg, D-06099 Halle (Saale), Germany
| | - Haitong Wan
- College of Life Science, Zhejiang Chinese Medical University, 548 Binwen Road, Hangzhou 310053, China
- Correspondence: (H.W.); (G.Z.)
| | - Guoying Zhou
- The Second Clinical Medical College, Zhejiang Chinese Medical University, 548 Binwen Road, Hangzhou 310053, China
- College of Life Science, Zhejiang Chinese Medical University, 548 Binwen Road, Hangzhou 310053, China
- Correspondence: (H.W.); (G.Z.)
| |
Collapse
|
20
|
Bray ER, Kirsner RS, Badiavas EV. Mesenchymal Stem Cell-Derived Extracellular Vesicles as an Advanced Therapy for Chronic Wounds. Cold Spring Harb Perspect Biol 2022; 14:a041227. [PMID: 35817513 PMCID: PMC9524280 DOI: 10.1101/cshperspect.a041227] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Chronic wounds are a significant challenge for patients, healthcare providers, and healthcare systems. Chronic wounds develop due to a complex interplay between chronic inflammation, tissue hypoxia, and oxidative stress, often occurring in the setting of advancing age. Ideally, new therapeutics should address all the components of chronic wound pathophysiology. Mesenchymal stem cell (MSC) therapies show significant promise to promote healing of chronic wounds. Extracellular vesicles (EVs) secreted by MSCs mediate many of their beneficial effects. We review the evidence demonstrating that MSC-EVs target the processes leading to chronic wounds. Additionally, we discuss how MSCs can be influenced to generate more potent wound healing EVs. Finally, we highlight the current state of EV clinical trials for wound healing and important preclinical studies that will lead to optimal use of MSC-EVs for patient care.
Collapse
Affiliation(s)
- Eric R Bray
- Phillip Frost Department of Dermatology and Cutaneous Surgery
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida 33136, USA
| | | | - Evangelos V Badiavas
- Phillip Frost Department of Dermatology and Cutaneous Surgery
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida 33136, USA
| |
Collapse
|
21
|
miR-100-5p Promotes Epidermal Stem Cell Proliferation through Targeting MTMR3 to Activate PIP3/AKT and ERK Signaling Pathways. Stem Cells Int 2022; 2022:1474273. [PMID: 36045954 PMCID: PMC9421352 DOI: 10.1155/2022/1474273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 07/22/2022] [Accepted: 08/02/2022] [Indexed: 12/04/2022] Open
Abstract
Skin epidermal stem cells (EpSCs) play a critical role in wound healing and are ideal seed cells for skin tissue engineering. Exosomes from human adipose-derived stem cells (ADSC-Exos) promote human EpSC proliferation, but the underlying mechanism remains unclear. Here, we investigated the effect of miR-100-5p, one of the most abundant miRNAs in ADSC-Exos, on the proliferation of human EpSCs and explored the mechanisms involved. MTT and BrdU incorporation assays showed that miR-100-5p mimic transfection promoted EpSC proliferation in a time-dependent manner. Cell cycle analysis showed that miR-100-5p mimic transfection significantly decreased the percentage of cells in the G1 phase and increased the percentage of cells in the G2/M phase. Myotubularin-related protein 3 (MTMR3), a lipid phosphatase, was identified as a direct target of miR-100-5p. Knockdown of MTMR3 in EpSCs by RNA interference significantly enhanced cell proliferation, decreased the percentage of cells in the G1 phase and increased the percentage of cells in the S phase. Overexpression of MTMR3 reversed the proproliferative effect of miR-100-5p on EpSCs, indicating that miR-100-5p promoted EpSC proliferation by downregulating MTMR3. Mechanistic studies showed that transfection of EpSCs with miR-100-5p mimics elevated the intracellular PIP3 level, induced AKT and ERK phosphorylation, and upregulated cyclin D1, E1, and A2 expression, which could be attenuated by MTMR3 overexpression. Consistently, intradermal injection of ADSC-Exos or miR-100-5p-enriched ADSC-Exos into cultured human skin tissues significantly reduced MTMR3 expression and increased the thickness of the epidermis and the number of EpSCs in the basal layer of the epidermis. The aforementioned effect of miR-100-5p-enriched ADSC-Exos was stronger than that of ADSC-Exos and was reversed by MTMR3 overexpression. Collectively, our findings indicate that miR-100-5p promotes EpSC proliferation through MTMR3-mediated elevation of PIP3 and activation of AKT and ERK. miR-100-5p-enriched ADSC-Exos can be used to treat skin wound and expand EpSCs for generating epidermal autografts and engineered skin equivalents.
Collapse
|
22
|
Lv H, Liu H, Sun T, Wang H, Zhang X, Xu W. Exosome derived from stem cell: A promising therapeutics for wound healing. Front Pharmacol 2022; 13:957771. [PMID: 36003496 PMCID: PMC9395204 DOI: 10.3389/fphar.2022.957771] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 06/27/2022] [Indexed: 11/13/2022] Open
Abstract
A wound occurs when the epidermis and dermis of the skin are damaged internally and externally. The traditional wound healing method is unsatisfactory, which will prolong the treatment time and increase the treatment cost, which brings economic and psychological burdens to patients. Therefore, there is an urgent need for a new method to accelerate wound healing. As a cell-free therapy, exosome derived from stem cell (EdSC) offers new possibilities for wound healing. EdSC is the smallest extracellular vesicle secreted by stem cells with diameters of 30-150 nm and a lipid bilayer structure. Previous studies have found that EdSC can participate in and promote almost all stages of wound healing, including regulating inflammatory cells; improving activation of fibroblasts, keratinocytes, and endothelial cells; and adjusting the ratio of collagen Ⅰ and Ⅲ. We reviewed the relevant knowledge of wounds; summarized the biogenesis, isolation, and identification of exosomes; and clarified the pharmacological role of exosomes in promoting wound healing. This review provides knowledge support for the pharmacological study of exosomes.
Collapse
Affiliation(s)
| | | | | | | | | | - Wei Xu
- Department of Clinical Pharmacy, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
| |
Collapse
|
23
|
Free-cell therapeutics and mechanism of exosomes from adipose-derived stem cells in promoting wound healing: current understanding and future applications. Chin Med J (Engl) 2022; 135:1803-1805. [PMID: 35633586 PMCID: PMC9521765 DOI: 10.1097/cm9.0000000000001857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
|
24
|
Stem Cell-Derived Exosomes: A New Method for Reversing Skin Aging. Tissue Eng Regen Med 2022; 19:961-968. [PMID: 35809187 DOI: 10.1007/s13770-022-00461-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 04/06/2022] [Accepted: 04/13/2022] [Indexed: 10/17/2022] Open
Abstract
Senescence is an inevitable natural life process that involves structural and functional degeneration of tissues and organs. Recently, the process of skin aging has attracted much attention. Determining a means to delay or even reverse skin aging has become a research hotspot in medical cosmetology and anti-aging. Dysfunction in the epidermis and fibroblasts and changes in the composition and content of the extracellular matrix are common pathophysiological manifestations of skin aging. Reactive oxygen species and matrix metalloproteinases play essential roles in this process. Stem cells are pluripotent cells that possess self-replication abilities and can differentiate into multiple functional cells under certain conditions. These cells also possess a strong ability to facilitate tissue repair and regeneration. Stem cell transplantation has the potential for application in anti-aging therapy. Increasing studies have demonstrated that stem cells perform functions through paracrine processes, particularly those involving exosomes. Exosomes are nano-vesicular substances secreted by stem cells that participate in cell-to-cell communication by transporting their contents into target cells. In this chapter, the biological characteristics of exosomes were reviewed, including their effects on extracellular matrix formation, epidermal cell function, fibroblast function and antioxidation. Exosomes derived from stem cells may provide a new means to reverse skin aging.
Collapse
|
25
|
Chen J, Liu R, Huang T, Sun H, Jiang H. Adipose stem cells-released extracellular vesicles as a next-generation cargo delivery vehicles: a survey of minimal information implementation, mass production and functional modification. Stem Cell Res Ther 2022; 13:182. [PMID: 35505389 PMCID: PMC9062865 DOI: 10.1186/s13287-022-02849-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 04/12/2022] [Indexed: 02/07/2023] Open
Abstract
OBJECTIVES To investigate current situation of minimal information implementation highlighted by minimal information for studies of extracellular vesicles 2018 (MISEV2018) guidelines, and explore technological advances towards mass production and functional modification in aesthetic, plastic and reconstructive surgery. METHODS Original articles on extracellular vesicles (EVs) of adipose stem cells (ASCs) were identified. Statistics upon minimal information for EVs research, such as species, cell types, culture conditions, conditioned media harvesting parameters, EVs isolation/storage/identification/quantification, functional uptake and working concentration, were analyzed. RESULTS The items of cell culture conditions such as passage number, seeding density, conditioned media harvesting time, functional uptake and working concentration were poorly documented, with a reporting percentage of 47.13%, 54.02%, 29.89%, 62.07% and 36.21%, respectively. However, there were some studies not reporting information of ASCs origin, culture medium, serum, EVs isolation methods, quantification and identification of EVs, accounting for 3.45%, 10.34%, 6.90%, 3.45%, 18.39% and 4.02%, respectively. Serum deprivation and trophic factors stimuli were attempted for EVs mass production. Several technological advances towards functional modification included hypoxia pre-condition, engineering EVs and controlled release. Presently, ASCs EVs have been applied in multiple fields, including diabetic/non-diabetic wound healing, angiogenesis, inflammation modulation, fat grafting, hair regeneration, antiaging, and healing and regeneration of cartilage/bone/peripheral nerve/tendon. CONCLUSION Our results highlight normative reporting of ASCs EVs in functional studies to increase reliability and reproducibility of scientific publications. The advances towards mass production and functional modification of ASCs EVs are also recommended to enhance therapeutic effects.
Collapse
Affiliation(s)
- Jianguo Chen
- Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 33 Badachu Road, Shijingshan District, Beijing, 100144, People's Republic of China
| | - Ruiquan Liu
- Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 33 Badachu Road, Shijingshan District, Beijing, 100144, People's Republic of China
| | - Tianyu Huang
- Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 33 Badachu Road, Shijingshan District, Beijing, 100144, People's Republic of China
| | - Hengyun Sun
- Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 33 Badachu Road, Shijingshan District, Beijing, 100144, People's Republic of China
| | - Haiyue Jiang
- Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 33 Badachu Road, Shijingshan District, Beijing, 100144, People's Republic of China.
| |
Collapse
|
26
|
Prasai A, Jay JW, Jupiter D, Wolf SE, El Ayadi A. Role of Exosomes in Dermal Wound Healing: A Systematic Review. J Invest Dermatol 2022; 142:662-678.e8. [PMID: 34461128 PMCID: PMC9400548 DOI: 10.1016/j.jid.2021.07.167] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 07/08/2021] [Accepted: 07/27/2021] [Indexed: 12/22/2022]
Abstract
Cell-based therapy imparts its therapeutic effects through soluble GFs and vesicular bodies such as exosomes. A systematic review with a meta-analysis of preclinical studies was conducted according to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses and the modified Stroke Therapy Academic Industry Roundtable guidelines to identify exosomes as an archetype biological therapy for dermal wound healing and to provide guidelines for the concentrations to be used in preclinical studies. A total of 51 rodent studies were included in the systematic review and 9 were included in the meta-analysis section. Three independent reviewers cross-screened eligibility and selected studies for quality assessment from 3,064 published studies on exosomes and wound healing. The mean quality scores for all studies were 5.08 ± 0.752 and 5.11 ± 1.13 for systematic review and meta-analysis, respectively. Exosome effects were reported to have the highest efficacy at 7 days (OR = 1.82, 95% confidence interval = 0.69‒2.95) than at 14 days (OR = 2.29, 95% confidence interval = 0.01‒4.56) after administration. Exosomes were reported to regulate all phases of skin wound healing, mostly by the actions of circulating microRNA. The outcome of this review may be used to guide preclinical and clinical studies on the role of exosomes in wound healing.
Collapse
Affiliation(s)
- Anesh Prasai
- Department of Surgery, University of Texas Medical Branch, Galveston, Texas, USA.
| | | | | | | | | |
Collapse
|
27
|
Zhang Y, Li Y, Wang Q, Zheng D, Feng X, Zhao W, Cai L, Zhang Q, Xu H, Fu H. Attenuation of hepatic ischemia‑reperfusion injury by adipose stem cell‑derived exosome treatment via ERK1/2 and GSK‑3β signaling pathways. Int J Mol Med 2022; 49:13. [PMID: 34878156 PMCID: PMC8711591 DOI: 10.3892/ijmm.2021.5068] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 10/14/2021] [Indexed: 11/23/2022] Open
Abstract
Exosomes are an emerging therapeutic tool for the treatment of tissue injuries. In the present study, the protective effect of isolated exosomes from adipose‑derived stem cells (ADSCs‑exo) against hepatic ischemia‑reperfusion (I/R) injury was explored. Hepatic I/R injury was achieved by inducing ischemia for 60 min followed by reperfusion for 2 and 6 h. Pre‑treatment with ADSCs‑exo revealed a significant reduction in necrosis and apoptosis in liver tissue induced by I/R injury. Hypoxic oxidative stress was managed by exosome‑mediated reduced reactive oxygen species and increased superoxide dismutase that in turn protected mitochondrial damage and apoptosis. Reduction in inflammatory mediators such as IL‑1β and TNF‑α was also observed and protection of hepatocytes from I/R injury was evidenced by a significant decrease in biochemical markers of liver damage (alanine transaminase, aspartate transaminase and lactate dehydrogenase). Exosomal prostaglandin E2 (PGE2)‑mediated ERK1/2 and GSK‑3β phosphorylation were revealed to increase Bcl‑2 and decrease Bax expression with mitochondrial permeability transition pore‑inhibition which may be considered a prime mechanism of exosome‑mediated hepatoprotection. In conclusion, our results indicated that ADSCs‑exo pre‑treatment is effective in protecting liver I/R injury.
Collapse
Affiliation(s)
- Yaqing Zhang
- Department of Anesthesiology, Changzheng Hospital, Naval Medical University, Shanghai 200003, P.R. China
| | - Yonghua Li
- Department of Anesthesiology, Changzheng Hospital, Naval Medical University, Shanghai 200003, P.R. China
| | - Qilong Wang
- Department of Anesthesiology, Changzheng Hospital, Naval Medical University, Shanghai 200003, P.R. China
| | - Dongyu Zheng
- Department of Anesthesiology, Changzheng Hospital, Naval Medical University, Shanghai 200003, P.R. China
| | - Xue Feng
- Department of Liver Surgery and Liver Transplantation Center, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P.R. China
| | - Wei Zhao
- Department of Obstetrics and Gynecology, Inner Mongolia Autonomous Region Corps Hospital of Chinese People's Armed Police Force, Hohhot, Inner Mongolia Autonomous Region 010040, P.R. China
| | - Linlin Cai
- Department of Anesthesiology, Wuxi People's Hospital, Wuxi, Jiangsu 214023, P.R. China
| | - Qingqing Zhang
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, P.R. China
| | - Haitao Xu
- Department of Anesthesiology, Changzheng Hospital, Naval Medical University, Shanghai 200003, P.R. China
| | - Hailong Fu
- Department of Anesthesiology, Changzheng Hospital, Naval Medical University, Shanghai 200003, P.R. China
| |
Collapse
|
28
|
Xie Y, Tian S. Exosomal MicroRNA-325 Enhances Trophoblast Migration and Invasion Through Downregulation of Lethal-7b and Upregulation of Forkhead Box Protein O1 Expression in Preeclampsia. J BIOMATER TISS ENG 2022. [DOI: 10.1166/jbt.2022.2902] [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
We aimed to explore the mechanism by how microRNA (miRNA)-325 derived from marrow mesenchymal stem cell exosomes (MSC-exos) affects the trophoblast progression in preeclampsia (PE). RT-qPCR detected the level of miRNA let-7b and FOXO1 in the placenta tissue of PE patients. Functional
experiment was performed to analyze the effect of FOXO1 inhibitor and let-7b mimics on cell migration, invasion and apoptosis through Transwell assay and TUNEL staining. The trophoblast cell was co-cultured with overexpressed-miR-325 MSC-exos to measure gene expression and cell progression.
let-7b was highly and FOXO1 was lowly expressed in PE placenta tissue. let-7b directly targeted and inhibited FOXO1 expression. Importantly, as miR-325 was internalized by trophoblast cells through MSC-exos, MSC-exos overexpressing miR-325 inhibited let-7b expression in trophoblasts, up-regulated
FOXO1 and activated AKT signaling pathway. Further, MSC-exos treatment promoted invasion and migration of trophoblast cell and inhibited apoptosis. In conclusion, miR-325 derived from MSC-exos promotes the invasion and migration of trophoblast cells in PE through inhibition of let7b and upregulation
of FOXO1.
Collapse
Affiliation(s)
- Ying Xie
- Department of Obstetrics, Huangshi Maternity & Children’s Health Hospital, Edong Healthcare Group, Women’s & Children’s Hospital Affiliated to Hubei Polytechnic University, Huangshi, Hubei, 435000, China
| | - Shan Tian
- Department of Obstetrics, Huangshi Maternity & Children’s Health Hospital, Edong Healthcare Group, Women’s & Children’s Hospital Affiliated to Hubei Polytechnic University, Huangshi, Hubei, 435000, China
| |
Collapse
|
29
|
Liu Y, Liu Y, Wu M, Zou R, Mao S, Cong P, Hou M, Jin H, Zhao Y, Bao Y. Adipose-derived mesenchymal stem cell-loaded β-chitin nanofiber hydrogel promote wound healing in rats. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2022; 33:12. [PMID: 35050422 PMCID: PMC8776676 DOI: 10.1007/s10856-021-06630-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Accepted: 11/28/2021] [Indexed: 05/14/2023]
Abstract
Because of stem cells are limited by the low efficiency of their cell homing and survival in vivo, cell delivery systems and scaffolds have attracted a great deal of attention for stem cells' successful clinical practice. β-chitin nanofibers (β-ChNF) were prepared from squid pens in this study. Fourier transform infrared spectroscopy, X-ray diffraction and scanning electron microscopy proved that β-ChNFs with the diameter of 5 to 10 nm were prepared. β-ChNF dispersion became gelled upon the addition of cell culture medium. Cell culture experiments showed that β-ChNFs exhibited negligible cytotoxicity towards ADSCs and L929 cells, and it was found that more exosomes were secreted by the globular ADSCs grown in the β-ChNF hydrogel. The vivo experiments of rats showed that the ADSCs-loaded β-ChNF hydrogel could directly cover the wound surface and significantly accelerate the wound healing and promote the generation of epithelization, granulation tissue and collagen. In addition, the ADSCs-loaded β-ChNF hydrogel clearly regulated the expressions of VEGFR, α-SMA, collagen I and collagen III. Finally, we showed that ADSCs-loaded β-ChNF hydrogel activated the TGFβ/smad signaling. The neutralization of TGFβ markedly reduced Smad phosphorylation and the expressions of TIMP1, VEGFR and α-SMA. Taken together, these findings suggest that ADSCs-loaded β-ChNF hydrogel promises for treating wounds that are challenge to heal via conventional methods. Graphical abstract.
Collapse
Affiliation(s)
- Ying Liu
- National Engineering Laboratory for Druggable Gene and Protein Screening, Northeast Normal University, Changchun, 130117, China
- Emergency Medicine Department of General Hospital of Northern Theater Command, Laboratory of Rescue Center of Severe Wound and Trauma PLA, Shenyang, 110016, China
| | - Yunen Liu
- Emergency Medicine Department of General Hospital of Northern Theater Command, Laboratory of Rescue Center of Severe Wound and Trauma PLA, Shenyang, 110016, China
| | - Mi Wu
- Jihua Laboratory, Foshan, 528200, China
| | - Rufei Zou
- Jihua Laboratory, Foshan, 528200, China
| | - Shun Mao
- Emergency Medicine Department of General Hospital of Northern Theater Command, Laboratory of Rescue Center of Severe Wound and Trauma PLA, Shenyang, 110016, China
| | - Peifang Cong
- Emergency Medicine Department of General Hospital of Northern Theater Command, Laboratory of Rescue Center of Severe Wound and Trauma PLA, Shenyang, 110016, China
| | - Mingxiao Hou
- Emergency Medicine Department of General Hospital of Northern Theater Command, Laboratory of Rescue Center of Severe Wound and Trauma PLA, Shenyang, 110016, China
| | - Hongxu Jin
- Emergency Medicine Department of General Hospital of Northern Theater Command, Laboratory of Rescue Center of Severe Wound and Trauma PLA, Shenyang, 110016, China.
| | - Yan Zhao
- Jihua Laboratory, Foshan, 528200, China.
| | - Yongli Bao
- National Engineering Laboratory for Druggable Gene and Protein Screening, Northeast Normal University, Changchun, 130117, China.
| |
Collapse
|
30
|
Bray ER, Oropallo AR, Grande DA, Kirsner RS, Badiavas EV. Extracellular Vesicles as Therapeutic Tools for the Treatment of Chronic Wounds. Pharmaceutics 2021; 13:1543. [PMID: 34683836 PMCID: PMC8541217 DOI: 10.3390/pharmaceutics13101543] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 09/16/2021] [Accepted: 09/17/2021] [Indexed: 12/17/2022] Open
Abstract
Chronic wounds develop when the orderly process of cutaneous wound healing is delayed or disrupted. Development of a chronic wound is associated with significant morbidity and financial burden to the individual and health-care system. Therefore, new therapeutic modalities are needed to address this serious condition. Mesenchymal stem cells (MSCs) promote skin repair, but their clinical use has been limited due to technical challenges. Extracellular vesicles (EVs) are particles released by cells that carry bioactive molecules (lipids, proteins, and nucleic acids) and regulate intercellular communication. EVs (exosomes, microvesicles, and apoptotic bodies) mediate key therapeutic effects of MSCs. In this review we examine the experimental data establishing a role for EVs in wound healing. Then, we explore techniques for designing EVs to function as a targeted drug delivery system and how EVs can be incorporated into biomaterials to produce a personalized wound dressing. Finally, we discuss the status of clinically deploying EVs as a therapeutic agent in wound care.
Collapse
Affiliation(s)
- Eric R. Bray
- Phillip Frost Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (E.R.B.); (R.S.K.)
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Alisha R. Oropallo
- Comprehensive Wound Healing Center and Hyperbarics, Department of Vascular Surgery, Donald and Barbara Zucker School of Medicine, Hofstra/Northwell Health, Hempstead, NY 11549, USA; (A.R.O.); (D.A.G.)
- Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY 11030, USA
| | - Daniel A. Grande
- Comprehensive Wound Healing Center and Hyperbarics, Department of Vascular Surgery, Donald and Barbara Zucker School of Medicine, Hofstra/Northwell Health, Hempstead, NY 11549, USA; (A.R.O.); (D.A.G.)
- Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY 11030, USA
- Department of Orthopedic Surgery, Long Island Jewish Medical Center, Northwell Health, New Hyde Park, NY 11040, USA
| | - Robert S. Kirsner
- Phillip Frost Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (E.R.B.); (R.S.K.)
| | - Evangelos V. Badiavas
- Phillip Frost Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (E.R.B.); (R.S.K.)
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| |
Collapse
|
31
|
Extracellular Vesicles in Skin Wound Healing. Pharmaceuticals (Basel) 2021; 14:ph14080811. [PMID: 34451909 PMCID: PMC8400229 DOI: 10.3390/ph14080811] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 08/13/2021] [Accepted: 08/16/2021] [Indexed: 12/13/2022] Open
Abstract
Each year, millions of individuals suffer from a non-healing wound, abnormal scarring, or injuries accompanied by an infection. For these cases, scientists are searching for new therapeutic interventions, from which one of the most promising is the use of extracellular vesicles (EVs). Naturally, EV-based signaling takes part in all four wound healing phases: hemostasis, inflammation, proliferation, and remodeling. Such an extensive involvement of EVs suggests exploiting their action to modulate the impaired healing phase. Furthermore, next to their natural wound healing capacity, EVs can be engineered for better defined pharmaceutical purposes, such as carrying specific cargo or targeting specific destinations by labelling them with certain surface proteins. This review aims to promote scientific awareness in basic and translational research of EVs by summarizing the current knowledge about their natural role in each stage of skin repair and the most recent findings in application areas, such as wound healing, skin regeneration, and treatment of dermal diseases, including the stem cell-derived, plant-derived, and engineered EVs.
Collapse
|
32
|
Zeng QL, Liu DW. Mesenchymal stem cell-derived exosomes: An emerging therapeutic strategy for normal and chronic wound healing. World J Clin Cases 2021; 9:6218-6233. [PMID: 34434989 PMCID: PMC8362559 DOI: 10.12998/wjcc.v9.i22.6218] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 04/08/2021] [Accepted: 06/22/2021] [Indexed: 02/06/2023] Open
Abstract
Skin wound healing is a complex biological process. Mesenchymal stem cells (MSCs) play an important role in skin wound repair due to their multidirectional differentiation potential, hematopoietic support, promotion of stem cell implantation, self-replication, and immune regulation. Exosomes are vesicles with diameters of 40-100 nm that contain nucleic acids, proteins, and lipids and often act as mediators of cell-to-cell communication. Currently, many clinical scientists have carried out cell-free therapy for skin wounds, especially chronic wounds, using exosomes derived from MSCs. This review focuses on the latest research progress on the mechanisms of action associated with the treatment of wound healing with exosomes derived from different MSCs, the latest research progress on the combination of exosomes and other biological or nonbiological factors for the treatment of chronic skin wounds, and the new prospects and development goals of cell-free therapy.
Collapse
Affiliation(s)
- Qin-Lu Zeng
- Burns Institute, The First Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi Province, China
- First Clinical Medical College, Nanchang University, Nanchang 330006, Jiangxi Province, China
| | - De-Wu Liu
- Burns Institute, The First Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi Province, China
| |
Collapse
|
33
|
Weiliang Z, Lili G. Research Advances in the Application of Adipose-Derived Stem Cells Derived Exosomes in Cutaneous Wound Healing. Ann Dermatol 2021; 33:309-317. [PMID: 34341631 PMCID: PMC8273313 DOI: 10.5021/ad.2021.33.4.309] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 12/21/2020] [Accepted: 01/11/2021] [Indexed: 12/15/2022] Open
Abstract
Cutaneous wound healing has always been an intractable medical problem for both clinicians and researchers, with an urgent need for more efficacious methods to achieve optimal outcomes morphologically and functionally. Stem cells, the body's rapid response 'road repair crew,' being on standby to combat tissue injuries, are an essential part of regenerative medicine. Currently, the use of adipose-derived stem cells (ADSCs), a kind of mesenchymal stem cells with multipotent differentiation and self-renewal capacity, is surging in the field of cutaneous wound healing. ADSCs may exert influences either by releasing paracrine signalling factors or differentiating into mature adipose cells to provide the 'building blocks' for engineered tissue. As an important paracrine substance released from ADSCs, exosomes are a kind of extracellular vesicles and carrying various bioactive molecules mediating adjacent or distant intercellular communication. Previous studies have indicated that ADSCs derived exosomes (ADSCs-Exos) promoted skin wound healing by affecting all stages of wound healing, including regulating inflammatory response, promoting proliferation and migration of fibroblasts or keratinocytes, facilitating angiogenesis, and regulating remodeling of extracellular matrix, which have provided new opportunities for understanding how ADSCs-Exos mediate intercellular communication in pathological processes of the skin and therapeutic strategies for cutaneous wound repair. In this review, we focus on elucidating the role of ADSCs-Exos at various stages of cutaneous wound healing, detailing the latest developments, and presenting some challenges necessary to be addressed in this field, with the expectation of providing a new perspective on how to best utilize this powerful cell-free therapy in the future.
Collapse
Affiliation(s)
- Zeng Weiliang
- Department of Cosmetic and Plastic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Guo Lili
- Department of Cosmetic and Plastic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
34
|
Dinescu S, Dobranici A, Tecucianu R, Selaru A, Balahura R, Ignat S, Costache M. Exosomes as Part of the Human Adipose-Derived Stem Cells Secretome- Opening New Perspectives for Cell-Free Regenerative Applications. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1312:139-163. [PMID: 32986128 DOI: 10.1007/5584_2020_588] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Human adipose-derived stem cells (hASCs) represent a great resource for regenerative medicine based on their accessibility, self-renewal potential, low immunogenicity, high proliferative rate and potential to differentiate on multiple lineages. Their secretome is rich in chemokines, cytokines and protein growth factors that are actively involved in regeneration processes. In addition, part of this secretome are also the exosomes (hASC-exos), which display high content in proteins, messenger RNAs (mRNAs) and non-coding RNAs (ncRNAs). Due to their content, exosomes promote tissue regeneration by different mechanisms, either by activating or inhibiting several signaling pathways involved in wound healing, extracellular matrix remodeling, immunomodulation, angiogenesis, anti-apoptotic activity and cell migration, proliferation and differentiation. The use of hASC-exos may provide an improved alternative to standard therapies used in regenerative medicine, as a cell-free new approach with multiple possibilities to be modulated according to the patient needs. This review offers an updated overview on the functions and applications of hASC-exos in all areas of tissue regeneration, aiming to highlight to the reader the benefits of using hASCs in modern tissue engineering and regenerative medicine applications.
Collapse
Affiliation(s)
- Sorina Dinescu
- Department of Biochemistry and Molecular Biology, University of Bucharest, Bucharest, Romania. .,The Research Institute of the University of Bucharest, Bucharest, Romania.
| | - Alexandra Dobranici
- Department of Biochemistry and Molecular Biology, University of Bucharest, Bucharest, Romania
| | - Ramona Tecucianu
- Department of Biochemistry and Molecular Biology, University of Bucharest, Bucharest, Romania
| | - Aida Selaru
- Department of Biochemistry and Molecular Biology, University of Bucharest, Bucharest, Romania.,Victor Babes National Institute of Pathology, Bucharest, Romania
| | - Roxana Balahura
- Department of Biochemistry and Molecular Biology, University of Bucharest, Bucharest, Romania.,Victor Babes National Institute of Pathology, Bucharest, Romania
| | - Simona Ignat
- Department of Biochemistry and Molecular Biology, University of Bucharest, Bucharest, Romania
| | - Marieta Costache
- Department of Biochemistry and Molecular Biology, University of Bucharest, Bucharest, Romania.,The Research Institute of the University of Bucharest, Bucharest, Romania
| |
Collapse
|
35
|
Liu SC, Bamodu OA, Kuo KT, Fong IH, Lin CC, Yeh CT, Chen SG. Adipose-derived stem cell induced-tissue repair or wound healing is mediated by the concomitant upregulation of miR-21 and miR-29b expression and activation of the AKT signaling pathway. Arch Biochem Biophys 2021; 705:108895. [PMID: 33933426 DOI: 10.1016/j.abb.2021.108895] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 04/22/2021] [Accepted: 04/25/2021] [Indexed: 12/21/2022]
Abstract
BACKGROUND Adipose-derived stem cells (ADSCs), a subpopulation of mesenchymal stem cells, are characterized by their potential to differentiate into multiple cell lineages. Due to their abundance and relative ease of procurement, ADSCs are widely used for tissue repair and regeneration. However, the molecular mechanisms of the therapeutic effect of ADSCs remain unknown. METHODS MicroRNAs have emerged as important signaling molecules in skin wound healing, and their roles in ADSC-based therapies must be addressed. Here, we investigated the potential of ADSCs in improving cutaneous wound healing in vitro and in vivo. RESULTS We simulated the microenvironment of the wound site by coculturing human dermal fibroblasts (HDFs) with ADSCs. We found that cocultured HDFs expressed significantly higher levels of miR-29b and miR-21 and had higher proliferation and migration rates than ADSCs cultured without HDFs. Moreover, increased expression of Collagen Type I Alpha 1 Chain (COL1A1), Collagen Type III Alpha 1 Chain (COL3A1), alpha-smooth muscle actin (α-SMA), vascular endothelial growth factor (VEGF), and Phosphoinositide 3-kinase (PI3K), p-Akt and decreased expression of Phosphatase and tensin homolog (PTEN) and matrix metalloproteinase (MMP)-1 was detected, suggesting extracellular remodeling and fibroblast activation and proliferation. We validated the in vitro results by using a rodent skin excisional wound model and implanted ADSC sheets in the wound. Compared with the controls, wounds implanted with ADSC sheets had significantly higher rates of wound-closure; increased expression of α-SMA, VEGF, PI3k, PTEN, COL1A1, and COL3A1; decreased expression of PTEN and MMP1; and upregulated levels of miR-29b and miR-21 in the skin. CONCLUSION In summary, we evidenced that ADSCs facilitate the increase in miR-29b and miR-21 levels and promote the activation and proliferation of dermal fibroblasts and extracellular matrix (ECM) remodeling, with the associated release of VEGF. Thus, the ADSC-mediated increase in microRNAs is essential in tissue repair and has a therapeutic potential in cutaneous wound healing.
Collapse
Affiliation(s)
- Shao-Cheng Liu
- Department of Otolaryngology-Head and Neck Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei City, 114, Taiwan.
| | - Oluwaseun Adebayo Bamodu
- Department of Medical Research & Education, Taipei Medical University - Shuang Ho Hospital, New Taipei City, 235, Taiwan; Department of Hematology and Oncology, Cancer Center, Taipei Medical University - Shuang Ho Hospital, New Taipei City, 235, Taiwan.
| | - Kuang-Tai Kuo
- Division of Thoracic Surgery, Department of Surgery, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan; Division of Thoracic Surgery, Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.
| | - Iat-Hang Fong
- Department of Medical Research & Education, Taipei Medical University - Shuang Ho Hospital, New Taipei City, 235, Taiwan; Department of Hematology and Oncology, Cancer Center, Taipei Medical University - Shuang Ho Hospital, New Taipei City, 235, Taiwan.
| | - Chih-Cheng Lin
- Department of Biotechnology and Pharmaceutical, Yuanpei University of Medical Technology, No. 306, Yuanpei Street, Hsinchu, Taiwan.
| | - Chi-Tai Yeh
- Department of Medical Research & Education, Taipei Medical University - Shuang Ho Hospital, New Taipei City, 235, Taiwan; Department of Hematology and Oncology, Cancer Center, Taipei Medical University - Shuang Ho Hospital, New Taipei City, 235, Taiwan; Department of Biotechnology and Pharmaceutical, Yuanpei University of Medical Technology, No. 306, Yuanpei Street, Hsinchu, Taiwan.
| | - Shyi-Gen Chen
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan; Division of Plastic and Reconstructive Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan.
| |
Collapse
|
36
|
Burmeister DM, Chu GCY, Chao T, Heard TC, Gómez BI, Sousse LE, Natesan S, Christy RJ. ASCs derived from burn patients are more prone to increased oxidative metabolism and reactive oxygen species upon passaging. Stem Cell Res Ther 2021; 12:270. [PMID: 33957963 PMCID: PMC8100366 DOI: 10.1186/s13287-021-02327-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 04/07/2021] [Indexed: 11/16/2022] Open
Abstract
Background Patients with severe burn injury (over 20% of the total body surface area) experience profound hypermetabolism which significantly prolongs wound healing. Adipose-derived stem cells (ASCs) have been proposed as an attractive solution for treating burn wounds, including the potential for autologous ASC expansion. While subcutaneous adipocytes display an altered metabolic profile post-burn, it is not known if this is the case with the stem cells associated with the adipose tissue. Methods ASCs were isolated from discarded burn skin of severely injured human subjects (BH, n = 6) and unburned subcutaneous adipose tissue of patients undergoing elective abdominoplasty (UH, n = 6) and were analyzed at passages 2, 4, and 6. Flow cytometry was used to quantify ASC cell surface markers CD90, CD105, and CD73. Mitochondrial abundance and reactive oxygen species (ROS) production were determined with MitoTracker Green and MitoSOX Red, respectively, while JC-10 Mitochondrial Membrane Potential Assays were also performed. Mitochondrial respiration and glycolysis were analyzed with a high-resolution respirometer (Seahorse XFe24 Analyzer). Results There was no difference in age between BH and UH (34 ± 6 and 41 ± 4 years, respectively, P = 0.49). While passage 2 ASCs had lower ASC marker expression than subsequent passages, there were no significant differences in the expression between BH and UH ASCs. Similarly, no differences in mitochondrial abundance or membrane potential were found amongst passages or groups. Two-way ANOVA showed a significant effect (P < 0.01) of passaging on mitochondrial ROS production, with increased ROS in BH ASCs at later passages. Oxidative phosphorylation capacities (leak and maximal respiration) increased significantly in BH ASCs (P = 0.035) but not UH ASCs. On the contrary, basal glycolysis significantly decreased in BH ASCs (P = 0.011) with subsequent passaging, but not UH ASCs. Conclusions In conclusion, ASCs from burned individuals become increasingly oxidative and less glycolytic upon passaging when compared to ASCs from unburned patients. This increase in oxidative capacities was associated with ROS production in later passages. While the autologous expansion of ASCs holds great promise for treating burned patients with limited donor sites, the potential negative consequences of using them require further investigation.
Collapse
Affiliation(s)
- David M Burmeister
- Department of Medicine, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA. .,United States Army Institute of Surgical Research, JBSA Fort Sam Houston, 3698 Chambers Pass, San Antonio, TX, USA.
| | - Grace Chu-Yuan Chu
- United States Army Institute of Surgical Research, JBSA Fort Sam Houston, 3698 Chambers Pass, San Antonio, TX, USA
| | - Tony Chao
- University of Texas Medical Branch, 301 University Blvd, Galveston, TX, 77555, USA
| | - Tiffany C Heard
- United States Army Institute of Surgical Research, JBSA Fort Sam Houston, 3698 Chambers Pass, San Antonio, TX, USA
| | - Belinda I Gómez
- United States Army Institute of Surgical Research, JBSA Fort Sam Houston, 3698 Chambers Pass, San Antonio, TX, USA
| | - Linda E Sousse
- United States Army Institute of Surgical Research, JBSA Fort Sam Houston, 3698 Chambers Pass, San Antonio, TX, USA
| | - Shanmugasundaram Natesan
- United States Army Institute of Surgical Research, JBSA Fort Sam Houston, 3698 Chambers Pass, San Antonio, TX, USA
| | - Robert J Christy
- United States Army Institute of Surgical Research, JBSA Fort Sam Houston, 3698 Chambers Pass, San Antonio, TX, USA
| |
Collapse
|