1
|
Ding X, Xie S, Zhang W, Zhu Y, Xu D, Xian S, Sun H, Guo X, Li Y, Lu J, Tong X, Huang R, Ji S, Xia Z. Current application of tissue-engineered dermal scaffolds mimicking the extracellular matrix microenvironment in wound healing. Regen Ther 2025; 28:371-382. [PMID: 39896445 PMCID: PMC11786805 DOI: 10.1016/j.reth.2024.12.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Revised: 12/09/2024] [Accepted: 12/26/2024] [Indexed: 02/04/2025] Open
Abstract
With the continuous advancement of materials science, cell biology, and biotechnology, tissue engineering has introduced novel solutions to traditional wound healing approaches, particularly demonstrating significant potential in addressing complex or non-healing wounds. One of the key technologies in this field, dermal scaffolds, serve as wound coverage materials that mimic the structural framework of the dermis. They primarily assume the function of extracellular matrix, providing space for cell attachment, migration, and proliferation, thus supporting cellular growth and regulating multiple biological processes in healing. Tissue engineering utilizes combinations of natural or synthetic scaffolds, seeded cells, or growth factors to induce distinct effects in angiogenesis, extracellular matrix deposition, and functional recovery. Therefore, various bioengineered dermal scaffolds hold significant potential for clinical translation in wound healing. This review outlines various extracellular matrix molecules utilized in the development of dermal scaffolds, emphasizes recent progress in cell- and growth factor-modified scaffolds, and discusses the challenges and future perspectives in this evolving field.
Collapse
Affiliation(s)
| | | | | | - Yushu Zhu
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, People's Republic of China, Research Unit of Key Techniques for Treatment of Burns and Combined Burns and Trauma Injury, Chinese Academy of Medical Sciences, No. 168 Changhai Road, Shanghai 200433, China
| | - Dayuan Xu
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, People's Republic of China, Research Unit of Key Techniques for Treatment of Burns and Combined Burns and Trauma Injury, Chinese Academy of Medical Sciences, No. 168 Changhai Road, Shanghai 200433, China
| | - Shuyuan Xian
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, People's Republic of China, Research Unit of Key Techniques for Treatment of Burns and Combined Burns and Trauma Injury, Chinese Academy of Medical Sciences, No. 168 Changhai Road, Shanghai 200433, China
| | - Hanlin Sun
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, People's Republic of China, Research Unit of Key Techniques for Treatment of Burns and Combined Burns and Trauma Injury, Chinese Academy of Medical Sciences, No. 168 Changhai Road, Shanghai 200433, China
| | - Xinya Guo
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, People's Republic of China, Research Unit of Key Techniques for Treatment of Burns and Combined Burns and Trauma Injury, Chinese Academy of Medical Sciences, No. 168 Changhai Road, Shanghai 200433, China
| | - Yixu Li
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, People's Republic of China, Research Unit of Key Techniques for Treatment of Burns and Combined Burns and Trauma Injury, Chinese Academy of Medical Sciences, No. 168 Changhai Road, Shanghai 200433, China
| | - Jianyu Lu
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, People's Republic of China, Research Unit of Key Techniques for Treatment of Burns and Combined Burns and Trauma Injury, Chinese Academy of Medical Sciences, No. 168 Changhai Road, Shanghai 200433, China
| | - Xirui Tong
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, People's Republic of China, Research Unit of Key Techniques for Treatment of Burns and Combined Burns and Trauma Injury, Chinese Academy of Medical Sciences, No. 168 Changhai Road, Shanghai 200433, China
| | - Runzhi Huang
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, People's Republic of China, Research Unit of Key Techniques for Treatment of Burns and Combined Burns and Trauma Injury, Chinese Academy of Medical Sciences, No. 168 Changhai Road, Shanghai 200433, China
| | - Shizhao Ji
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, People's Republic of China, Research Unit of Key Techniques for Treatment of Burns and Combined Burns and Trauma Injury, Chinese Academy of Medical Sciences, No. 168 Changhai Road, Shanghai 200433, China
| | - Zhaofan Xia
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, People's Republic of China, Research Unit of Key Techniques for Treatment of Burns and Combined Burns and Trauma Injury, Chinese Academy of Medical Sciences, No. 168 Changhai Road, Shanghai 200433, China
| |
Collapse
|
2
|
Alsaab HO, Almutairy B, Almobarki AO, Mughaedh MAA, Alzahrani MS. Exosome's role in ovarian disease pathogenesis and therapy; Focus on ovarian cancer and failure. J Reprod Immunol 2025; 167:104403. [PMID: 39662240 DOI: 10.1016/j.jri.2024.104403] [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: 10/28/2024] [Revised: 11/16/2024] [Accepted: 11/28/2024] [Indexed: 12/13/2024]
Abstract
In the eukaryotic system, exosomes are categorized as unique extracellular vesicles with dimensions ranging from 30 to 150 nm. These vesicles contain a variety of endogenous molecules, such as proteins, DNA, mRNA, microRNA, and circular RNA. They are essential for a wide range of metabolic events and have the potential to be used as therapeutic or diagnostic targets for a number of diseases, including ovarian diseases. By inducing changes in the surrounding environment, the donor exosomes transfer their contents to the receiving cells, so demonstrating the biological implications of major interactions between cells. Mesenchymal stem cells (MSCs) have produced exosomes have shown promise as a treatment for premature organ failure (POF or POI). Furthermore, exosomal transport has many complexities, and contributes to the pathophysiology of ovarian cancer by affecting cell growth, migration, metastastsis and etc. Owing to these facts, in this paper, we present the progress developed in the understanding of exosomes as a viable therapeutic avenue and indisputable prognostic targets in ovarian disorders.
Collapse
Affiliation(s)
- Hashem O Alsaab
- Department of Pharmaceutics and Pharmaceutical Technology, Taif University, Taif 21944, Saudi Arabia.
| | - Bandar Almutairy
- Department of Pharmacology, College of Pharmacy, Shaqra University, Shaqra 11961, Saudi Arabia.
| | | | | | - Mohammad S Alzahrani
- Department of Clinical Pharmacy, College of Pharmacy, Taif University, P.O. Box 11099, Taif , Saudi Arabia
| |
Collapse
|
3
|
Liu H, Ji M, Yang T, Zou S, Qiu X, Zhan F, Chen J, Yan F, Ding F, Li P. Regulation of fibroblast phenotype in osteoarthritis using CDKN1A-loaded copper sulfide nanoparticles delivered by mesenchymal stem cells. Am J Physiol Cell Physiol 2025; 328:C679-C698. [PMID: 39819042 DOI: 10.1152/ajpcell.00573.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 11/26/2024] [Accepted: 12/12/2024] [Indexed: 01/19/2025]
Abstract
This study aimed to investigate the regulation of fibroblast phenotypes by mesenchymal stem cells (MSCs) delivering copper sulfide (CuS) nanoparticles (NPs) loaded with CDKN1A plasmids and their role in cartilage repair during osteoarthritis (OA). Single-cell RNA sequencing data from the GEO database were analyzed to identify subpopulations within the OA immune microenvironment. Quality control, filtering, principal component analysis (PCA) dimensionality reduction, and tSNE clustering were performed to obtain detailed cell subtypes. Pseudotime analysis was used to understand the developmental trajectory of fibroblasts, and GO/KEGG enrichment analyses highlighted biological processes related to fibroblast function. Transcriptomic data and WGCNA identified CDKN1A as a key regulatory gene. A biomimetic CuS@CDKN1A nanosystem was constructed and loaded into MSCs to create MSCs@CuS@CDKN1A. The characterization of this system confirmed its efficient cellular uptake by fibroblasts. In vitro experiments demonstrated that MSCs@CuS@CDKN1A significantly modulated fibroblast phenotypes and improved the structure, proliferation, reduced apoptosis, and enhanced migration of IL-1β-stimulated chondrocytes. In vivo, an OA mouse model was treated with intra-articular injections of MSCs@CuS@CDKN1A. Micro-CT scans revealed a significant reduction in osteophyte formation and improved joint space compared with control groups. Histological analysis, including H&E, Safranin O-Fast Green, and toluidine blue staining, confirmed improved cartilage integrity, whereas the International Osteoarthritis Research Society (OARSI) scoring indicated reduced disease severity. Immunofluorescence showed upregulated CDKN1A expression, decreased MMP13, and reduced α-SMA expression in fibroblast subtypes. Major organs exhibited no signs of toxicity, confirming the biocompatibility and safety of the treatment. These findings suggest that MSCs@CuS@CDKN1A can effectively regulate fibroblast activity and promote cartilage repair, providing a promising therapeutic strategy for OA treatment.NEW & NOTEWORTHY This study introduces MSCs@CuS@CDKN1A, a nanoengineered MSC platform that targets fibroblast phenotypes in osteoarthritis (OA). By modulating CDKN1A expression, this innovative approach not only enhances cartilage repair but also effectively mitigates fibroblast-driven inflammation, marking a significant advancement in OA therapeutics with demonstrated efficacy and biocompatibility.
Collapse
Affiliation(s)
- Hong Liu
- Department of Orthopedics, Chongqing University Three Gorges Hospital, Chongqing, People's Republic of China
- Chongqing Municipality Clinical Research Center for Geriatric Diseases, Chongqing, People's Republic of China
| | - Ming Ji
- Department of Orthopedics, Chongqing University Three Gorges Hospital, Chongqing, People's Republic of China
- Chongqing Municipality Clinical Research Center for Geriatric Diseases, Chongqing, People's Republic of China
| | - Tao Yang
- Department of Orthopedics, Chongqing University Three Gorges Hospital, Chongqing, People's Republic of China
- Chongqing Municipality Clinical Research Center for Geriatric Diseases, Chongqing, People's Republic of China
| | - Shihua Zou
- Department of Orthopedics, Chongqing University Three Gorges Hospital, Chongqing, People's Republic of China
- Chongqing Municipality Clinical Research Center for Geriatric Diseases, Chongqing, People's Republic of China
| | - Xingan Qiu
- Department of Orthopedics, Chongqing University Three Gorges Hospital, Chongqing, People's Republic of China
- Chongqing Municipality Clinical Research Center for Geriatric Diseases, Chongqing, People's Republic of China
| | - Fangbiao Zhan
- Department of Orthopedics, Chongqing University Three Gorges Hospital, Chongqing, People's Republic of China
- Chongqing Municipality Clinical Research Center for Geriatric Diseases, Chongqing, People's Republic of China
- School of Medicine, Chongqing University, Chongqing, People's Republic of China
| | - Jian Chen
- Department of Orthopedics, Chongqing University Three Gorges Hospital, Chongqing, People's Republic of China
- Chongqing Municipality Clinical Research Center for Geriatric Diseases, Chongqing, People's Republic of China
- School of Medicine, Chongqing University, Chongqing, People's Republic of China
| | - Fei Yan
- Chongqing Municipality Clinical Research Center for Geriatric Diseases, Chongqing, People's Republic of China
- School of Medicine, Chongqing University, Chongqing, People's Republic of China
| | - Fan Ding
- Department of Orthopedics, General Hospital of Central Theater Command, Wuhan, People's Republic of China
| | - Ping Li
- Division of Orthopedics, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, People's Republic of China
| |
Collapse
|
4
|
Abdallah S, Tabebi M, Qanadilo S, Ali N, Wang J, D'Arcy P, Zhong W, Sjoberg F, Elmasry M, El-Serafi A. Modulation of biological activities in adipose derived stem cells by histone deacetylation. Sci Rep 2025; 15:3629. [PMID: 39880862 PMCID: PMC11779964 DOI: 10.1038/s41598-024-84652-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 12/25/2024] [Indexed: 01/31/2025] Open
Abstract
Difficult-to-heal wounds management accounts for about 4% of healthcare costs, highlighting the need for innovative solutions. Extracellular signals drive cell proliferation during tissue regeneration, while epigenetic mechanisms regulate stem cell homeostasis, differentiation, and skin repair. Exploring epigenetic regulation in adipose-derived stem cells (ADSCs) holds promise for improving skin injury treatments. We investigated the effects of histone deacetylase inhibitor (SAHA) on ADSCs to better understand its cellular and molecular impacts. ADSCs were treated with SAHA for 72 h, showing no change in cell viability at the studied concentrations. However, the expression of histone deacetylase decreased at 1000 nM, while the cell proliferation marker Ki-67 increased after SAHA treatment, as confirmed by immunofluorescence. CCND1 gene expression increased, whereas protein expression of the proliferating cell nuclear antigen (PCNA) decreased. Cell cycle analysis showed an increase in G2 phase in SAHA-treated cells. Microarray analysis revealed 74 upregulated and 40 downregulated differentially expressed genes, including upregulation of P53 targets, CDKN1A and MDM2. Proteomic analysis identified 631 upregulated and 823 downregulated proteins compared to the vehicle. Pathway enrichment analysis showed cell cycle, ATP-dependent chromatin remodeling and DNA processes were among the affected pathways. This study suggests SAHA modulates ADSCs' biological processes, highlighting its potential for skin regeneration.
Collapse
Affiliation(s)
- Sallam Abdallah
- The Department of Biomedical and Clinical Sciences (BKV), Linköping University, Linköping, Sweden
| | - Mouna Tabebi
- The Department of Biomedical and Clinical Sciences (BKV), Linköping University, Linköping, Sweden
- Clinical Genomics Linköping, Linköping University, Linköping, Sweden
| | - Sawsan Qanadilo
- Department of Biological Sciences, The University of Jordan, Amman, Jordan
| | - Neserin Ali
- Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Jing Wang
- The Department of Biomedical and Clinical Sciences (BKV), Linköping University, Linköping, Sweden
| | - Pádraig D'Arcy
- The Department of Biomedical and Clinical Sciences (BKV), Linköping University, Linköping, Sweden
| | - Wen Zhong
- The Department of Biomedical and Clinical Sciences (BKV), Linköping University, Linköping, Sweden
| | - Folke Sjoberg
- The Department of Biomedical and Clinical Sciences (BKV), Linköping University, Linköping, Sweden
| | - Moustafa Elmasry
- The Department of Biomedical and Clinical Sciences (BKV), Linköping University, Linköping, Sweden
- Department of Hand Surgery and Plastic Surgery and Burns, University Hospital, Linköping, Sweden
| | - Ahmed El-Serafi
- The Department of Biomedical and Clinical Sciences (BKV), Linköping University, Linköping, Sweden.
- Department of Hand Surgery and Plastic Surgery and Burns, University Hospital, Linköping, Sweden.
| |
Collapse
|
5
|
Zhao S, Kong H, Qi D, Qiao Y, Li Y, Cao Z, Wang H, He X, Liu H, Yang H, Gao S, Liu T, Xie J. Epidermal stem cell derived exosomes-induced dedifferentiation of myofibroblasts inhibits scarring via the miR-203a-3p/PIK3CA axis. J Nanobiotechnology 2025; 23:56. [PMID: 39881312 PMCID: PMC11776291 DOI: 10.1186/s12951-025-03157-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Accepted: 01/22/2025] [Indexed: 01/31/2025] Open
Abstract
Hypertrophic scar (HS) is a common fibroproliferative disorders with no fully effective treatments. The conversion of fibroblasts to myofibroblasts is known to play a critical role in HS formation, making it essential to identify molecules that promote myofibroblast dedifferentiation and to elucidate their underlying mechanisms. In this study, we used comparative transcriptomics and single-cell sequencing to identify key molecules and pathways that mediate fibrosis and myofibroblast transdifferentiation. Epidermal stem cell-derived extracellular vesicles (EpiSC-EVs) were isolated via ultracentrifugation and filtration, followed by miRNA sequencing to identify miRNAs targeting key molecules. After in vitro and in vivo treatment with EpiSC-EVs, we assessed antifibrotic effects through scratch assays, collagen contraction assays, Western blotting, and immunofluorescence. Transcriptomic sequencing and rescue experiments were used to investigate the molecular mechanism by which miR-203a-3p in EpiSC-EVs induces myofibroblast dedifferentiation. Our results indicate that PIK3CA is overexpressed in HS tissues and positively correlates with fibrosis. EpiSC-EVs were absorbed by scar-derived fibroblasts, promoting dedifferentiation from myofibroblasts to quiescent fibroblasts. Mechanistically, miR-203a-3p in EpiSC-EVs plays an essential role in inhibiting PIK3CA expression and PI3K/AKT/mTOR pathway hyperactivation, thereby reducing scar formation. In vivo studies confirmed that EpiSC-EVs attenuate excessive scarring through the miR-203a-3p/PIK3CA axis, suggesting EpiSC-EVs as a promising therapeutic approach for HS.
Collapse
Affiliation(s)
- Shixin Zhao
- Department of Traumatic Orthopedics, Henan Provincial People's Hospital & The People's Hospital of Zhengzhou University, No. 7 Weiwu Road, Zhengzhou, Henan, 450003, China
- Henan Orthopedics Research Institute, No. 7 Weiwu Road, Zhengzhou, Henan, 450003, China
- Department of Burns, Wound Repair and Reconstruction, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China
| | - Haoran Kong
- Department of Traumatic Orthopedics, Henan Provincial People's Hospital & The People's Hospital of Zhengzhou University, No. 7 Weiwu Road, Zhengzhou, Henan, 450003, China
- Henan Orthopedics Research Institute, No. 7 Weiwu Road, Zhengzhou, Henan, 450003, China
| | - Dahu Qi
- Department of Traumatic Orthopedics, Henan Provincial People's Hospital & The People's Hospital of Zhengzhou University, No. 7 Weiwu Road, Zhengzhou, Henan, 450003, China
- Henan Orthopedics Research Institute, No. 7 Weiwu Road, Zhengzhou, Henan, 450003, China
| | - Yushuang Qiao
- Department of Traumatic Orthopedics, Henan Provincial People's Hospital & The People's Hospital of Zhengzhou University, No. 7 Weiwu Road, Zhengzhou, Henan, 450003, China
- Henan Orthopedics Research Institute, No. 7 Weiwu Road, Zhengzhou, Henan, 450003, China
| | - Yu Li
- Department of Traumatic Orthopedics, Henan Provincial People's Hospital & The People's Hospital of Zhengzhou University, No. 7 Weiwu Road, Zhengzhou, Henan, 450003, China
- Henan Orthopedics Research Institute, No. 7 Weiwu Road, Zhengzhou, Henan, 450003, China
| | - Zhiming Cao
- Department of Traumatic Orthopedics, Henan Provincial People's Hospital & The People's Hospital of Zhengzhou University, No. 7 Weiwu Road, Zhengzhou, Henan, 450003, China
- Henan Orthopedics Research Institute, No. 7 Weiwu Road, Zhengzhou, Henan, 450003, China
| | - Hanwen Wang
- Department of Burns, Wound Repair and Reconstruction, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China
| | - Xuefeng He
- Department of Burns, Wound Repair and Reconstruction, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China
| | - Hengdeng Liu
- Department of Burns, Wound Repair and Reconstruction, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China
| | - Hao Yang
- Department of Burns, Wound Repair and Reconstruction, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China
| | - Suyue Gao
- Department of Burns, Wound Repair and Reconstruction, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China
| | - Tao Liu
- Department of Traumatic Orthopedics, Henan Provincial People's Hospital & The People's Hospital of Zhengzhou University, No. 7 Weiwu Road, Zhengzhou, Henan, 450003, China.
- Henan Orthopedics Research Institute, No. 7 Weiwu Road, Zhengzhou, Henan, 450003, China.
| | - Julin Xie
- Department of Burns, Wound Repair and Reconstruction, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China.
| |
Collapse
|
6
|
Li X, Cui J, Ning LJ, Hu RN, Zhao LL, Luo JJ, Xie XY, Zhang YJ, Luo JC, Li ZY, Qin TW. Response of a tenomodulin-positive subpopulation of human adipose-derived stem cells to decellularized tendon slices. Biomed Mater 2025; 20:025004. [PMID: 39746323 DOI: 10.1088/1748-605x/ada509] [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: 10/22/2024] [Accepted: 01/02/2025] [Indexed: 01/04/2025]
Abstract
The selection of appropriate cell sources is vital for the regeneration and repair of tendons using stem cell-based approaches. Human adipose-derived stem cells (hADSCs) have emerged as a promising therapeutic strategy for tendon injuries. However, the heterogeneity of hADSCs can lead to inconsistent or suboptimal therapeutic outcomes. In this study, we isolated and identified a tenomodulin (TNMD)-positive subpopulation from hADSCs (TNMD+hADSCs) using flow cytometry and then assessed the cellular response of this subpopulation to decellularized tendon slices (DTSs), including cell proliferation, migration, and tenogenic differentiation, using the CCK-8 assay, transwell migration assay, and quantitative real-time polymerase chain reaction. Our findings revealed that TNMD+hADSCs maintained the general characteristics of stem cells and exhibited significantly higher expressions of tendon-related markers compared to hADSCs. Importantly, DTSs significantly enhanced the proliferation, migration, and tenogenic differentiation of TNMD+hADSCs. This study provides preliminary experimental evidence for the translational application of ADSCs for tendon regeneration and repair.
Collapse
Affiliation(s)
- Xuan Li
- Department of Orthopedic Surgery and Orthopedic Research Institute, Stem Cells and Tissue Engineering Research Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Jing Cui
- Department of Orthopedic Surgery and Orthopedic Research Institute, Stem Cells and Tissue Engineering Research Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Liang-Ju Ning
- Department of Orthopedic Surgery and Orthopedic Research Institute, Stem Cells and Tissue Engineering Research Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Ruo-Nan Hu
- Department of Orthopedic Surgery and Orthopedic Research Institute, Stem Cells and Tissue Engineering Research Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Lei-Lei Zhao
- Department of Orthopedic Surgery and Orthopedic Research Institute, Stem Cells and Tissue Engineering Research Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Jia-Jiao Luo
- Department of Orthopedic Surgery and Orthopedic Research Institute, Stem Cells and Tissue Engineering Research Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Xin-Yue Xie
- Department of Orthopedic Surgery and Orthopedic Research Institute, Stem Cells and Tissue Engineering Research Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Yan-Jing Zhang
- Department of Orthopedic Surgery and Orthopedic Research Institute, Stem Cells and Tissue Engineering Research Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Jing-Cong Luo
- Department of Orthopedic Surgery and Orthopedic Research Institute, Stem Cells and Tissue Engineering Research Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Zheng-Yong Li
- Department of Burn and Plastic Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Ting-Wu Qin
- Department of Orthopedic Surgery and Orthopedic Research Institute, Stem Cells and Tissue Engineering Research Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| |
Collapse
|
7
|
Li P, Cao L, Liu T, Lu X, Ma Y, Wang H. The Effect of Adipose-Derived Stem Cell (ADSC)-Exos on the Healing of Autologous Skin Grafts in Miniature Pigs. Int J Mol Sci 2025; 26:479. [PMID: 39859193 PMCID: PMC11764972 DOI: 10.3390/ijms26020479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 12/06/2024] [Accepted: 01/06/2025] [Indexed: 01/27/2025] Open
Abstract
The skin functions as the body's primary defense barrier; when compromised, it can lead to dehydration, infection, shock, or potentially life-threatening conditions. Miniature pigs exhibit skin characteristics and healing processes highly analogous to humans. Mesenchymal stem cells contribute to skin injury repair through a paracrine mechanism involving exosomes. This research examines whether adipose-derived MSC exosomes effectively enhance healing following autologous skin grafting in miniature pigs. It also compares the roles and distinctions of ADSCs and ADSC-Exos in inflammatory responses and tissue regeneration. This study found significantly reduced levels of oxidative stress products and pro-inflammatory factors, while antioxidant factors, anti-inflammatory factors, and pro-regenerative factors were elevated, and anti-regenerative factor levels decreased. Moreover, the expression levels of key markers-namely, PI3K, Akt, and mTOR-in the regeneration-associated signaling pathway were increased. The alterations in these indicators indicate that ADSC-Exos can regulate inflammatory responses and promote regeneration. This study provides a novel theoretical foundation for the implementation of acellular therapy in clinical settings.
Collapse
Affiliation(s)
- Pujun Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150300, China; (P.L.); (L.C.); (T.L.); (X.L.); (Y.M.)
- Heilongjiang Provincial Key Laboratory of Pathogenic Mechanism for Animal Disease and Comparative Medicine, Harbin 150300, China
| | - Lei Cao
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150300, China; (P.L.); (L.C.); (T.L.); (X.L.); (Y.M.)
- Heilongjiang Provincial Key Laboratory of Pathogenic Mechanism for Animal Disease and Comparative Medicine, Harbin 150300, China
| | - Tao Liu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150300, China; (P.L.); (L.C.); (T.L.); (X.L.); (Y.M.)
- Heilongjiang Provincial Key Laboratory of Pathogenic Mechanism for Animal Disease and Comparative Medicine, Harbin 150300, China
| | - Xiangyu Lu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150300, China; (P.L.); (L.C.); (T.L.); (X.L.); (Y.M.)
- Heilongjiang Provincial Key Laboratory of Pathogenic Mechanism for Animal Disease and Comparative Medicine, Harbin 150300, China
| | - Yajun Ma
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150300, China; (P.L.); (L.C.); (T.L.); (X.L.); (Y.M.)
- Heilongjiang Provincial Key Laboratory of Pathogenic Mechanism for Animal Disease and Comparative Medicine, Harbin 150300, China
| | - Hongbin Wang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150300, China; (P.L.); (L.C.); (T.L.); (X.L.); (Y.M.)
- Heilongjiang Provincial Key Laboratory of Pathogenic Mechanism for Animal Disease and Comparative Medicine, Harbin 150300, China
| |
Collapse
|
8
|
Dean J, Hoch C, Wollenberg B, Navidzadeh J, Maheta B, Mandava A, Knoedler S, Sherwani K, Baecher H, Schmitz A, Alfertshofer M, Heiland M, Kreutzer K, Koerdt S, Knoedler L. Advancements in bioengineered and autologous skin grafting techniques for skin reconstruction: a comprehensive review. Front Bioeng Biotechnol 2025; 12:1461328. [PMID: 39840132 PMCID: PMC11747595 DOI: 10.3389/fbioe.2024.1461328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 12/03/2024] [Indexed: 01/23/2025] Open
Abstract
The reconstruction of complex skin defects challenges clinical practice, with autologous skin grafts (ASGs) as the traditional choice due to their high graft take rate and patient compatibility. However, ASGs have limitations such as donor site morbidity, limited tissue availability, and the necessity for multiple surgeries in severe cases. Bioengineered skin grafts (BSGs) aim to address these drawbacks through advanced tissue engineering and biomaterial science. This study conducts a systematic review to describe the benefits and shortcomings of BSGs and ASGs across wound healing efficacy, tissue integration, immunogenicity, and functional outcomes focusing on wound re-epithelialization, graft survival, and overall aesthetic outcomes. Preliminary findings suggest ASGs show superior early results, while BSGs demonstrate comparable long-term outcomes with reduced donor site morbidity. This comparative analysis enhances understanding of bioengineered alternatives in skin reconstruction, potentially redefining best practices based on efficacy, safety, and patient-centric outcomes, highlighting the need for further innovation in bioengineered solutions.
Collapse
Affiliation(s)
- Jillian Dean
- School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Cosima Hoch
- Department of Otolaryngology, Head and Neck Surgery, School of Medicine and Health, Technical University of Munich (TUM), Munich, Germany
| | - Barbara Wollenberg
- Department of Otolaryngology, Head and Neck Surgery, School of Medicine and Health, Technical University of Munich (TUM), Munich, Germany
| | - Justin Navidzadeh
- School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Bhagvat Maheta
- California Northstate University College of Medicine, Elk Grove, CA, United States
| | - Anisha Mandava
- School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Samuel Knoedler
- Institute of Regenerative Biology and Medicine, Helmholtz Zentrum München, Munich, Germany
| | - Khalil Sherwani
- Institute of Regenerative Biology and Medicine, Helmholtz Zentrum München, Munich, Germany
| | - Helena Baecher
- Institute of Regenerative Biology and Medicine, Helmholtz Zentrum München, Munich, Germany
| | - Alina Schmitz
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Department of Oral and Maxillofacial Surgery, Berlin, Germany
| | - Michael Alfertshofer
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Department of Oral and Maxillofacial Surgery, Berlin, Germany
| | - Max Heiland
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Department of Oral and Maxillofacial Surgery, Berlin, Germany
| | - Kilian Kreutzer
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Department of Oral and Maxillofacial Surgery, Berlin, Germany
| | - Steffen Koerdt
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Department of Oral and Maxillofacial Surgery, Berlin, Germany
| | - Leonard Knoedler
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Department of Oral and Maxillofacial Surgery, Berlin, Germany
| |
Collapse
|
9
|
Huang Z, Niu R, Xu Q, Zhang R, Hu W, Qin Y, Wang X, Xu Q, Xia Y, Fan Y, Lu C. Impact of Maternal BPA Exposure during Pregnancy on Obesity in Male Offspring: A Mechanistic Mouse Study of Adipose-Derived Exosomal miRNA. ENVIRONMENTAL HEALTH PERSPECTIVES 2025; 133:17011. [PMID: 39886984 PMCID: PMC11783688 DOI: 10.1289/ehp14888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 12/18/2024] [Accepted: 12/26/2024] [Indexed: 02/01/2025]
Abstract
BACKGROUND The widespread use of bisphenol A (BPA) has led to universal exposure among the population, raising concerns about its health effects. Epidemiological studies have linked environmentally relevant levels of BPA exposure to obesity. OBJECTIVES We aimed to uncover the complex mechanisms by which oral exposure during pregnancy with BPA affects the offspring. METHODS We conducted a two-stage mouse study. In stage 1, we gavaged dams with BPA at 0.05, 0.5, and 5 mg / kg per day during pregnancy, and we tracked the offspring's weight and diet to 12 wk of age. In stage 2, exosomes from BPA-exposed dams and offspring were injected into pregnant mice and 3-wk-old males, respectively, and the mice were observed up to 12 wk. We then sequenced exosomal microRNAs (miRNAs) in male offspring whose dams had been exposed to BPA during pregnancy and checked their expression in adipose, liver, and serum samples at weeks 3, 6, 9, and 12. Finally, we explored the functions of exosomes and exosomal miRNAs secreted by adipose-derived mesenchymal stem cells, and we investigated whether the exosomes and miRNAs they secreted could affect glucose uptake, triglyceride synthesis, and the expression of genes related to glucose and lipid metabolism in alpha mouse liver 12 cells. RESULTS Gavage of 0.05 mg / kg per day of BPA during pregnancy in dams led to obesity in male offspring mice, and injection of exosomes from male offspring with BPA exposure during pregnancy also induced similar outcomes in the next generation of male pups. Exosomal miRNA sequencing identified differentially expressed miRNAs associated with BPA-induced obesity in male offspring, revealing sustained high expression of miRNAs in adipose tissue and a gradual increase in the liver and serum over time. Further mechanistic studies showed that exosomes derived from BPA-treated adipose-derived stem cells reduced the expression of peroxisome proliferator-activated receptor-gamma and fibroblast growth factor 21, leading to impaired insulin signaling and lipid metabolism in hepatocytes. Overexpression of miR-124-3p in hepatocytes mimicked these effects; in contrast, knockdown of miR-124-3p or inhibition of exosome secretion reversed them. DISCUSSION The present study corroborates the regulatory function of adipose-derived exosomal miRNAs in obesity in male offspring mice resulting from BPA exposure during pregnancy. Exosomal miRNA may be a key and novel molecular biomarker in the adverse effects of chemical exposure during pregnancy. https://doi.org/10.1289/EHP14888.
Collapse
Affiliation(s)
- Zhenyao Huang
- Key Laboratory of Human Genetics and Environmental Medicine, School of Public Health, Xuzhou Medical University, Xuzhou, China
| | - Rui Niu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
- Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Qiaoqiao Xu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
- Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Rui Zhang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
- Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Weiyue Hu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
- Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Yufeng Qin
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
- Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Xinru Wang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
- Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Qiujin Xu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
- Chinese Research Academy of Environmental Sciences, Beijing, China
| | - Yankai Xia
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
- Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Yun Fan
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
- Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Chuncheng Lu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
- Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China
| |
Collapse
|
10
|
Zou X, Xu H, Qian W. Macrophage Polarization in the Osteoarthritis Pathogenesis and Treatment. Orthop Surg 2025; 17:22-35. [PMID: 39638774 PMCID: PMC11735378 DOI: 10.1111/os.14302] [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: 06/08/2024] [Revised: 11/04/2024] [Accepted: 11/07/2024] [Indexed: 12/07/2024] Open
Abstract
Osteoarthritis (OA) is a prevalent degenerative disorder that severely impacts quality of life due to pain and disability. Although the pathophysiology of OA remains incompletely understood, recent research highlights the role of synovial inflammation in OA onset and progression, driven primarily by inflammatory infiltrates, especially macrophages, in the synovium. These macrophages respond to the local microenvironment, polarizing into either pro-inflammatory (M1) or anti-inflammatory (M2) subtypes. This review focuses on the role of macrophage polarization in OA pathogenesis and treatment, emphasizing how M1/M2 polarization is influenced by pathways such as STAT, NF-κB, caspase, and MAPK. These pathways induce low-grade inflammation within OA-affected joints, altering chondrocyte metabolism, inhibiting cartilage repair, and impairing mesenchymal stem cell chondrogenesis, thereby contributing to OA progression. Additionally, this review discusses potential therapies targeting macrophage polarization, encompassing compounds, proteins, cells, and microRNAs, to offer insights into novel treatment strategies for OA.
Collapse
Affiliation(s)
- Xiongfei Zou
- Department of Orthopedic SurgeryPeking Union Medical College HospitalBeijingChina
| | - Hongjun Xu
- Department of Orthopedic SurgeryPeking Union Medical College HospitalBeijingChina
| | - Wenwei Qian
- Department of Orthopedic SurgeryPeking Union Medical College HospitalBeijingChina
| |
Collapse
|
11
|
Hou B, Li C, Yang F, Deng W, Hu C, Liu C, Chen Y, Xiao X, Huang X, Deng J, Xie S. Ultrasmall Antioxidant Copper Nanozyme to Enhance Stem Cell Microenvironment for Promoting Diabetic Wound Healing. Int J Nanomedicine 2024; 19:13563-13578. [PMID: 39720217 PMCID: PMC11668326 DOI: 10.2147/ijn.s487647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 12/13/2024] [Indexed: 12/26/2024] Open
Abstract
Purpose Stem cell therapy is a promising approach for treating chronic diabetic wounds. However, its effectiveness is significantly limited by the high oxidative stress environment and persistent inflammation induced by diabetes. Strategies to overcome these challenges are essential to enhance the therapeutic potential of stem cell therapy. Methods Cu5.4O ultrasmall nanoparticles (Cu5.4O-USNPs), known for their excellent reactive oxygen species (ROS) scavenging properties, were utilized to protect adipose-derived stem cells (ADSCs) from oxidative stress injury. In vitro experiments were conducted to evaluate the viability, paracrine activity, and anti-inflammatory capabilities of ADSCs loaded with Cu5.4O-USNPs under oxidative stress conditions. In vivo experiments in diabetic mice were performed to assess the therapeutic effects of Cu5.4O-USNP-loaded ADSCs on wound healing, including their impact on inflammation, collagen synthesis, angiogenesis, and wound closure. Results ADSCs treated with Cu5.4O-USNPs showed significantly enhanced viability, paracrine activity, and anti-inflammatory properties under oxidative stress conditions in vitro. In diabetic mice, Cu5.4O-USNP-loaded ADSCs reduced inflammatory responses in wound tissues, promoted collagen synthesis and angiogenesis, and accelerated diabetic wound healing. These findings suggest that Cu5.4O-USNPs effectively mitigate the adverse effects of oxidative stress and inflammation, enhancing the therapeutic efficacy of ADSCs. Conclusion This study presents a simple and effective approach to improve the therapeutic potential of stem cell therapy for diabetic wounds. By incorporating Cu5.4O-USNPs, the antioxidative and anti-inflammatory capabilities of ADSCs are significantly enhanced, offering a promising strategy for ROS-related tissue repair and chronic wound healing.
Collapse
Affiliation(s)
- Biao Hou
- Department of Hand and Foot Microsurgery, The Affiliated Nanhua Hospital, Hengyang Medical College, University of South China, Hengyang, Hunan, People’s Republic of China
| | - Chengyuan Li
- Department of Pathology, School of Basic Medicine, Central South University, Changsha, Hunan, People’s Republic of China
| | - Fen Yang
- Department of Infectious Diseases, The Affiliated Nanhua Hospital, Hengyang Medical College, University of South China, Hengyang, Hunan, People’s Republic of China
| | - Wanjun Deng
- Department of Hand and Foot Microsurgery, The Affiliated Nanhua Hospital, Hengyang Medical College, University of South China, Hengyang, Hunan, People’s Republic of China
| | - Chao Hu
- Department of Hand and Foot Microsurgery, The Affiliated Nanhua Hospital, Hengyang Medical College, University of South China, Hengyang, Hunan, People’s Republic of China
| | - Changxiong Liu
- Department of Hand and Foot Microsurgery, The Affiliated Nanhua Hospital, Hengyang Medical College, University of South China, Hengyang, Hunan, People’s Republic of China
| | - Yanming Chen
- Department of Hand and Foot Microsurgery, The Affiliated Nanhua Hospital, Hengyang Medical College, University of South China, Hengyang, Hunan, People’s Republic of China
| | - Xiangjun Xiao
- Department of Hand and Foot Microsurgery, The Affiliated Nanhua Hospital, Hengyang Medical College, University of South China, Hengyang, Hunan, People’s Republic of China
| | - Xiongjie Huang
- Department of Hand and Foot Microsurgery, The Affiliated Nanhua Hospital, Hengyang Medical College, University of South China, Hengyang, Hunan, People’s Republic of China
| | - Jun Deng
- Department of Hand and Foot Microsurgery, The Affiliated Nanhua Hospital, Hengyang Medical College, University of South China, Hengyang, Hunan, People’s Republic of China
- Institute of Burn Research, Southwest Hospital, State Key Laboratory of Trauma, Burn and Combined Injury, Chongqing Key Laboratory for Disease Proteomics, Army Medical University, Chongqing, People’s Republic of China
| | - Songlin Xie
- Department of Hand and Foot Microsurgery, The Affiliated Nanhua Hospital, Hengyang Medical College, University of South China, Hengyang, Hunan, People’s Republic of China
| |
Collapse
|
12
|
Wang YW, Tan PC, Li QF, Xu XW, Zhou SB. Adipose tissue protects against skin photodamage through CD151- and AdipoQ- EVs. Cell Commun Signal 2024; 22:594. [PMID: 39696450 DOI: 10.1186/s12964-024-01978-z] [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: 10/13/2024] [Accepted: 12/03/2024] [Indexed: 12/20/2024] Open
Abstract
To clarify the protective effects of subcutaneous adipose tissue (SAT) against photodamage, we utilized nude mouse skin with or without SAT. Skin and fibroblasts were treated with adipose tissue-derived extracellular vesicles (AT-EVs) or extracellular vesicles derived from adipose-derived stem cells (ADSC-EVs) to demonstrate that SAT protects the overlying skin from photodamage primarily through AT-EVs. Surprisingly, AT-EVs stimulated fibroblast proliferation more rapidly than ADSC-EVs did. The yield of AT-EVs from the same volume of AT was 200 times greater than that of ADSC-EVs. To compare the differences between AT-EVs and ADSC-EVs, we used a proximity barcoding assay (PBA) to analyze the surface proteins on individual particles of these two types of EVs. PBA analysis revealed that AT-EVs contain diverse subpopulations, with 83.42% expressing CD151, compared to only 1.98% of ADSC-EVs. Furthermore, AT-EVs are internalized more rapidly by cells than ADSC-EVs, as our study demonstrated that CD151-positive AT-EVs were endocytosed more quickly than their CD151-negative counterparts. Additionally, adiponectin in AT-EVs activated the AMPK pathway and inhibited the NF-κB pathway, enhancing fibroblast protection against photodamage. The significantly higher yield and faster acquisition of AT-EVs compared to ADSC-EVs underscore their potential for broader applications.
Collapse
Affiliation(s)
- Yan-Wen Wang
- Department of Plastic & Reconstructive Surgery, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Plastic and Burn Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Poh-Ching Tan
- Department of Plastic & Reconstructive Surgery, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qing-Feng Li
- Department of Plastic & Reconstructive Surgery, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Xue-Wen Xu
- Department of Plastic and Burn Surgery, West China Hospital, Sichuan University, Chengdu, China.
| | - Shuang-Bai Zhou
- Department of Plastic & Reconstructive Surgery, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
13
|
Hekimoglu ER, Esrefoglu M, Cimen FBK, Pasin Ö, Dedeakayogullari H. Therapeutic Potential of Stromal Vascular Fraction in Enhancing Wound Healing: A Preclinical Study. Aesthetic Plast Surg 2024:10.1007/s00266-024-04554-5. [PMID: 39681692 DOI: 10.1007/s00266-024-04554-5] [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: 03/16/2024] [Accepted: 11/11/2024] [Indexed: 12/18/2024]
Abstract
BACKGROUND Adipose tissue provides an abundant source of stromal vascular fraction (SVF) cells for immediate administration. It can also give rise to many multipotent adipose-derived stromal cells. SVF is the population of cells obtained from mechanical or enzymatic digestion of lipoaspirate with no necessity for cell culture or expansion. Recently, the heterogeneous cell population found in the SVF gained wide-ranging translational significance in regenerative medicine. METHODS Forty-eight male rats were randomly divided into two main groups, including the control and SVF groups. Each group was further divided into four groups as follows: 0th-, 3rd-, 7th-, and 10th-day groups. A skin excision of 1 × 1 cm covering the epidermis and dermis was performed on the back skin. Just after the wound was created, a subepidermal injection of SVF was applied. SVF was obtained from human adipose tissue using Lipocube SVFTM. On the 0th (1 h after the injections), 3rd, 7th, and 10th days, rats were killed, and skin excisions from the wound areas tissues were performed. Histopathological, biochemical, and western blotting analyses were performed on tissues. RESULTS Our data showed that SVF obtained from a healthy woman improved wound healing in healthy rats. SVF has promoted wound healing mainly because of its antioxidant, antiapoptotic, and fibroblast/myofibroblast stimulating effects. SVF stimulated collagen production and contraction of the wound lips, supporting the closure. CONCLUSIONS Our study provides additional data about the efficacy and pathophysiological and molecular mechanisms of the action of SVF on wound healing in healthy subjects. Our study is an experimental animal study. NO LEVEL ASSIGNED This journal requires that authors assign a level of evidence to each submission to which Evidence-Based Medicine rankings are applicable. This excludes Review Articles, Book Reviews, and manuscripts that concern Basic Science, Animal Studies, Cadaver Studies, and Experimental Studies. For a full description of these Evidence-Based Medicine ratings, please refer to the Table of Contents or the online Instructions to Authors www.springer.com/00266.
Collapse
Affiliation(s)
- Emine Rumeysa Hekimoglu
- Department of Histology and Embryology, Faculty of Medicine, Bezmialem Vakıf University, Fatih, Istanbul, Turkey.
| | - Mukaddes Esrefoglu
- Department of Histology and Embryology, Faculty of Medicine, Bezmialem Vakıf University, Fatih, Istanbul, Turkey
| | - Fatma Bedia Karakaya Cimen
- Department of Histology and Embryology, Faculty of Medicine, Bezmialem Vakıf University, Fatih, Istanbul, Turkey
| | - Özge Pasin
- Department of Biostatistics, Faculty of Medicine, Bezmialem Vakif University, Istanbul, Turkey
| | - Huri Dedeakayogullari
- Department of Medical Biochemistry, Faculty of Medicine, Istinye University, Istanbul, Turkey
| |
Collapse
|
14
|
Wang S, Gao D, Li M, Wang Q, Du X, Yuan S. Enhanced Wound Healing and Autogenesis Through Lentiviral Transfection of Adipose-Derived Stem Cells Combined with Dermal Substitute. Biomedicines 2024; 12:2844. [PMID: 39767750 PMCID: PMC11673073 DOI: 10.3390/biomedicines12122844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Revised: 12/08/2024] [Accepted: 12/12/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Burns and chronic ulcers may cause severe skin loss, leading to critical health issues like shock, infection, sepsis, and multiple organ failure. Effective healing of full-thickness wounds may be challenging, with traditional methods facing limitations due to tissue shortage, infection, and lack of structural support. METHODS This study explored the combined use of gene transfection and dermal substitutes to improve wound healing. We used the DGTM (genes: DNP63A, GRHL2, TFAP2A, and MYC) factors to transfect adipose-derived stem cells (ADSCs), inducing their differentiation into keratinocytes. These transfected ADSCs were then incorporated into Pelnac® dermal substitutes to enhance vascularization and cellular proliferation for better healing outcomes. RESULTS Gene transfer using DGTM factors successfully induced keratinocyte differentiation in ADSCs. The application of these differentiated cells with Pelnac® dermal substitute to dermal wounds in mice resulted in the formation of skin tissue with a normal epidermal layer and proper collagen organization. This method alleviates the tediousness of the multiple transfection steps in previous protocols and the safety issues caused by using viral transfection reagents directly on the wound. Additionally, the inclusion of dermal substitutes addressed the lack of collagen and elastic fibers, promoting the formation of tissue resembling healthy skin rather than scar tissue. CONCLUSION Integrating DGTM factor-transfected ADSCs with dermal substitutes represents a novel strategy for enhancing the healing of full-thickness wounds. Further research and clinical trials are warranted to optimize and validate this innovative approach for broader clinical applications.
Collapse
Affiliation(s)
- Shiqi Wang
- Department of Plastic Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210002, China; (S.W.); (D.G.); (M.L.)
- Department of Plastic Surgery, Jinling Hospital, Nanjing School of Clinical Medicine, Southern Medical University, Nanjing 210002, China;
| | - Dinghui Gao
- Department of Plastic Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210002, China; (S.W.); (D.G.); (M.L.)
| | - Mingyu Li
- Department of Plastic Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210002, China; (S.W.); (D.G.); (M.L.)
| | - Qian Wang
- Department of Plastic Surgery, Jinling Hospital, Nanjing School of Clinical Medicine, Southern Medical University, Nanjing 210002, China;
| | - Xuanyu Du
- Department of Plastic Surgery, Jinling Hospital, School of Medicine, Southeast University, Nanjing 210002, China;
| | - Siming Yuan
- Department of Plastic Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210002, China; (S.W.); (D.G.); (M.L.)
- Department of Plastic Surgery, Jinling Hospital, Nanjing School of Clinical Medicine, Southern Medical University, Nanjing 210002, China;
| |
Collapse
|
15
|
Razavi ZS, Farokhi S, Mahmoudvand G, Karimi-Rouzbahani A, Farasati-Far B, Tahmasebi-Ghorabi S, Pazoki-Toroudi H, Saadat-Fakhr M, Afkhami H. Stem cells and bio scaffolds for the treatment of cardiovascular diseases: new insights. Front Cell Dev Biol 2024; 12:1472103. [PMID: 39726717 PMCID: PMC11669526 DOI: 10.3389/fcell.2024.1472103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Accepted: 10/01/2024] [Indexed: 12/28/2024] Open
Abstract
Mortality and morbidity from cardiovascular diseases are common worldwide. In order to improve survival and quality of life for this patient population, extensive efforts are being made to establish effective therapeutic modalities. New treatment options are needed, it seems. In addition to treating cardiovascular diseases, cell therapy is one of the most promising medical platforms. One of the most effective therapeutic approaches in this area is stem cell therapy. In stem cell biology, multipotent stem cells and pluripotent stem cells are divided into two types. There is evidence that stem cell therapy could be used as a therapeutic approach for cardiovascular diseases based on multiple lines of evidence. The effectiveness of stem cell therapies in humans has been studied in several clinical trials. In spite of the challenges associated with stem cell therapy, it appears that resolving them may lead to stem cells being used in cardiovascular disease patients. This may be an effective therapeutic approach. By mounting these stem cells on biological scaffolds, their effect can be enhanced.
Collapse
Affiliation(s)
- Zahra Sadat Razavi
- Physiology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Simin Farokhi
- Student Research Committee, USERN Office, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Golnaz Mahmoudvand
- Student Research Committee, USERN Office, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Arian Karimi-Rouzbahani
- Student Research Committee, USERN Office, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Bahareh Farasati-Far
- Department of Chemistry, Iran University of Science and Technology, Tehran, Iran
| | - Samaneh Tahmasebi-Ghorabi
- Master of Health Education, Research Expert, Clinical Research Development Unit, Emam Khomeini Hospital, Ilam University of Medical Sciences, Ilam, Iran
| | | | - Masoud Saadat-Fakhr
- Faculty of Medicine, Tehran Medical Sciences Branch, Islamic Azad University, Tehran, Iran
| | - Hamed Afkhami
- Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran
- Nervous System Stem Cells Research Center, Semnan University of Medical Sciences, Semnan, Iran
- Department of Medical Microbiology, Faculty of Medicine, Shahed University, Tehran, Iran
| |
Collapse
|
16
|
Cao X, Wu X, Zhang Y, Qian X, Sun W, Zhao Y. Emerging biomedical technologies for scarless wound healing. Bioact Mater 2024; 42:449-477. [PMID: 39308549 PMCID: PMC11415838 DOI: 10.1016/j.bioactmat.2024.09.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 08/16/2024] [Accepted: 09/01/2024] [Indexed: 09/25/2024] Open
Abstract
Complete wound healing without scar formation has attracted increasing attention, prompting the development of various strategies to address this challenge. In clinical settings, there is a growing preference for emerging biomedical technologies that effectively manage fibrosis following skin injury, as they provide high efficacy, cost-effectiveness, and minimal side effects compared to invasive and costly surgical techniques. This review gives an overview of the latest developments in advanced biomedical technologies for scarless wound management. We first introduce the wound healing process and key mechanisms involved in scar formation. Subsequently, we explore common strategies for wound treatment, including their fabrication methods, superior performance and the latest research developments in this field. We then shift our focus to emerging biomedical technologies for scarless wound healing, detailing the mechanism of action, unique properties, and advanced practical applications of various biomedical technology-based therapies, such as cell therapy, drug therapy, biomaterial therapy, and synergistic therapy. Finally, we critically assess the shortcomings and potential applications of these biomedical technologies and therapeutic methods in the realm of scar treatment.
Collapse
Affiliation(s)
- Xinyue Cao
- Department of Otolaryngology Head and Neck Surgery, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Xiangyi Wu
- Department of Otolaryngology Head and Neck Surgery, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Yuanyuan Zhang
- Department of Otolaryngology Head and Neck Surgery, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Xiaoyun Qian
- Department of Otolaryngology Head and Neck Surgery, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Weijian Sun
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325035, China
| | - Yuanjin Zhao
- Department of Otolaryngology Head and Neck Surgery, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
- Shenzhen Research Institute, Southeast University, Shenzhen, 518071, China
| |
Collapse
|
17
|
Bugajska-Liedtke M, Fatyga N, Brzozowski A, Bajek A, Maj M. Anaesthetics reduce the viability of adipose-derived stem cells. Adipocyte 2024; 13:2351870. [PMID: 38779963 PMCID: PMC11123512 DOI: 10.1080/21623945.2024.2351870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Accepted: 04/27/2024] [Indexed: 05/25/2024] Open
Abstract
Adipose-derived stem cells (ADSCs) are characterized by their low immunogenicity and unique immunosuppressive properties, providing many opportunities for autologous transplantation in regenerative medicine and plastic surgery. These methods are characterized by low rejection rates and intense stimulation of tissue regeneration. However, procedures during which fat tissue is harvested occur under local anaesthesia. To better understand the effects and mechanisms of anaesthetic compounds in cosmetic and therapeutic procedures, the present study used a mixture of these compounds (0.1% epinephrine, 8.4% sodium bicarbonate, and 4% articaine) and examined their impact on a human adipose-derived stem cell line. The results showed anesthetics' negative, dose-dependent effect on cell viability and proliferation, especially during the first 24 h of incubation. After extending the exposure to 48 and 72 h of incubation, cells adapted to new culture conditions. In contrast, no significant changes were observed in immunophenotype, cell cycle progression, and apoptosis. The results obtained from this study provide information on the effect of the selected mixture of anaesthetics on the characteristics and function of ASC52telo cells. The undesirable changes in the metabolic activity of cells suggest the need to search for new drugs to harvest cells with unaltered properties and higher efficacy in aesthetic medicine treatments.
Collapse
Affiliation(s)
- Maria Bugajska-Liedtke
- Department of Tissue Engineering, Ludwik Rydygier Collegium Medicum, Nicolaus Copernicus University, Bydgoszcz, Poland
| | - Nadia Fatyga
- Department of Tissue Engineering, Ludwik Rydygier Collegium Medicum, Nicolaus Copernicus University, Bydgoszcz, Poland
| | - Aleksander Brzozowski
- Department of Tissue Engineering, Ludwik Rydygier Collegium Medicum, Nicolaus Copernicus University, Bydgoszcz, Poland
| | - Anna Bajek
- Department of Oncology, Ludwik Rydygier Collegium Medicum, Nicolaus Copernicus University, Bydgoszcz, Poland
| | - Małgorzata Maj
- Department of Tissue Engineering, Ludwik Rydygier Collegium Medicum, Nicolaus Copernicus University, Bydgoszcz, Poland
| |
Collapse
|
18
|
Jung H, Jung Y, Seo J, Bae Y, Kim HS, Jeong W. Roles of extracellular vesicles from mesenchymal stem cells in regeneration. Mol Cells 2024; 47:100151. [PMID: 39547584 DOI: 10.1016/j.mocell.2024.100151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 11/09/2024] [Accepted: 11/10/2024] [Indexed: 11/17/2024] Open
Abstract
Mesenchymal stem cells (MSCs) are highly valued in regenerative medicine due to their ability to self-renew and differentiate into various cell types. Their therapeutic benefits are primarily due to their paracrine effects, in particular through extracellular vesicles (EVs), which are related to intercellular communication. Recent advances in EV production and extraction technologies highlight the potential of MSC-derived EVs (MSC-EVs) in tissue engineering and regenerative medicine. MSC-EVs offer several advantages over traditional cell therapies, including reduced toxicity and immunogenicity compared with whole MSCs. EVs carrying functional molecules such as growth factors, cytokines, and miRNAs play beneficial roles in tissue repair, fibrosis treatment, and scar prevention by promoting angiogenesis, skin cell migration, proliferation, extracellular matrix remodeling, and reducing inflammation. Despite the potential of MSC-EVs, there are several limitations to their use, including variability in quality, the need for standardized methods, low yield, and concerns about the composition of EVs and the potential risks. Overall, MSC-EVs are a promising alternative to cell-based therapies, and ongoing studies aim to understand their actions and optimize their use for better clinical outcomes in wound healing and skin regeneration.
Collapse
Affiliation(s)
- Hyeseong Jung
- Department of Biomedical Science, Catholic Kwandong University, Gangneung 25601, Republic of Korea
| | - Yuyeon Jung
- Department of Dental Hygiene, Catholic Kwandong University, Gangneung 25601, Republic of Korea
| | - Junsik Seo
- Department of Biomedical Science, Catholic Kwandong University, Gangneung 25601, Republic of Korea
| | - Yeongju Bae
- Department of Biomedical Science, Catholic Kwandong University, Gangneung 25601, Republic of Korea; Research Center for Marine Bio-Food and Medicine, Catholic Kwandong University, Gangneung 25601, Republic of Korea
| | - Han-Soo Kim
- Department of Biomedical Science, Catholic Kwandong University, Gangneung 25601, Republic of Korea
| | - Wooyoung Jeong
- Department of Biomedical Science, Catholic Kwandong University, Gangneung 25601, Republic of Korea; Research Center for Marine Bio-Food and Medicine, Catholic Kwandong University, Gangneung 25601, Republic of Korea.
| |
Collapse
|
19
|
Moreira MS, Mota ME, Ariga SKK, Jaguar GC, Marques MM. Mesenchymal stem cell therapies evidence in the treatment of irradiated salivary glands: A scoping review. J Clin Exp Dent 2024; 16:e1547-e1554. [PMID: 39822783 PMCID: PMC11733895 DOI: 10.4317/jced.62242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Accepted: 11/08/2024] [Indexed: 01/19/2025] Open
Abstract
Background Radiotherapy is one of the main treatments for head and neck cancer; however, due to its non-selectivity the glandular tissue can be affected. This scoping review aimed to identify the evidence about mesenchymal stem cell therapies for irradiated salivary gland regeneration. Material and Methods Two independent reviewers performed a literature search in MEDLINE/PubMed, Scopus, and Web of Science. The inclusion criteria were: 1) studies evaluation regeneration of irradiated salivary glands by stem cell therapies (cell-based or cell-free), (2) in vivo studies. Results The search resulted in 13 included studies. In general, both therapies demonstrated increased salivary levels, with mucin and amylase increased and structural protection of acinar cells. The cell-free therapy based on labial glands stem cell extract demonstrated a higher number of parasympathetic nerves. Conclusions Stem cell therapies (cell-free and cell-based) appear promising strategies for recovering saliva production in patients presenting irradiation-induced hyposalivation, with positive results toward regeneration of the form and function of the glands. However, due to the scarcity and heterogenicity of these pre-clinical studies, it is not possible to indicate which is the more indicated therapy. Key words:Mesenchymal stem cells, extracellular vesicles, exosomes, salivary glands, stem cell biology, hyposalivation, radiotherapy.
Collapse
Affiliation(s)
- Maria Stella Moreira
- Department of Stomatology, A.C. Camargo Cancer Center, São Paulo, SP, Brazil
- Department of Stomatology, School of Dentistry, University of São Paulo, São Paulo, SP, Brazil
| | - Maria Emília Mota
- Department of Stomatology, School of Dentistry, University of São Paulo, São Paulo, SP, Brazil
| | - Suely Kunimi Kubo Ariga
- School of Medicine, Emergency Medicine Laboratory, University of São Paulo, São Paulo, SP, Brazil
| | | | - Márcia Martins Marques
- Department of Stomatology, School of Dentistry, University of São Paulo, São Paulo, SP, Brazil
- AALZ, Sigmund Freud University, Vienna, Austria
| |
Collapse
|
20
|
Wang W, Ren Y, Yu Q, Jiang L, Yu C, Yue Z, Wang Y, Lu J, Che P, Li J, Sun H. Biodegradable exosome-engineered hydrogels for the prevention of peritoneal adhesions via anti-oxidation and anti-inflammation. Mater Today Bio 2024; 29:101312. [PMID: 39525394 PMCID: PMC11550211 DOI: 10.1016/j.mtbio.2024.101312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 08/16/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024] Open
Abstract
Peritoneal adhesions (PA) are a common and severe complication after abdominal surgery, impacting millions of patients worldwide. The use of anti-adhesion materials as physical barriers is an effective strategy to prevent postoperative adhesions. However, the local inflammatory microenvironment exerts a significant impact on the efficacy of anti-adhesion therapies. In this study, an injectable hydrogel based on oxidized dextran/carboxymethyl chitosan (DCC) is designed and prepared. Furthermore, the DCC hydrogel is specifically engineered to load the adipose mesenchymal stem cells (ADSCs)-derived exosomes (Exos) for the treatment of PA. The prepared DCC hydrogel can act as the physical barrier via covering the irregular wound surface effectively. Moreover, it shows controlled degradation property, enabling the regulated release of Exos. The DCC hydrogel loaded Exos (DCC/Exo) system has high antioxidant capacity, and can effectively modulate the inflammatory microenvironments and diminish apoptosis. Notably, it promotes a polarization shift towards the M2-like phenotype in macrophages. The RNA-seq analysis confirms that the DCC/Exo system exhibits significant anti-inflammatory properties and promotes a reduction in collagen deposition. Consequently, the DCC/Exo system can inhibit peritoneal adhesions significantly in a mouse cecum-abdominal wall injury model. These results demonstrate the DCC/Exo is an ideal material for preventing postoperative adhesions.
Collapse
Affiliation(s)
- Weitong Wang
- Department of Basic Medical Sciences, North China University of Science and Technology, Tangshan, 063210, China
| | - Yuchen Ren
- Department of Basic Medical Sciences, North China University of Science and Technology, Tangshan, 063210, China
| | - Qingyu Yu
- Frontiers Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering, Ministry of Education, School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300350, China
| | - Lijie Jiang
- Department of Basic Medical Sciences, North China University of Science and Technology, Tangshan, 063210, China
| | - Chaojie Yu
- Frontiers Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering, Ministry of Education, School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300350, China
| | - Zhiwei Yue
- Department of Basic Medical Sciences, North China University of Science and Technology, Tangshan, 063210, China
| | - Yue Wang
- Department of Basic Medical Sciences, North China University of Science and Technology, Tangshan, 063210, China
| | - Jiajun Lu
- Department of Basic Medical Sciences, North China University of Science and Technology, Tangshan, 063210, China
| | - Pengcheng Che
- School of Nursing and Rehabilitation, North China University of Science and Technology, Tangshan, 063210, China
| | - Junjie Li
- Frontiers Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering, Ministry of Education, School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300350, China
| | - Hong Sun
- Department of Basic Medical Sciences, North China University of Science and Technology, Tangshan, 063210, China
| |
Collapse
|
21
|
Yang G, Liu L, Xiao L, Ke S, Yang H, Lu Q. Accelerated scarless wound healing by dynamical regulation of angiogenesis and inflammation with immobilized asiaticoside and magnesium ions in silk nanofiber hydrogels. J Mater Chem B 2024; 12:11670-11684. [PMID: 39380345 DOI: 10.1039/d4tb01584c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/10/2024]
Abstract
It remains a challenge to effectively regulate the complicated microenvironment during the wound healing process. The optimization of synergistic action of angiogenesis and inflammation is considered critical for quicker scarless wound regeneration. Here, the silk nanofiber (SNF) acts as a multifunctional carrier to load hydrophobic asiaticoside (AC) and hydrophilic Mg2+, and also serves as an element to assemble injectable hydrogels, forming a bioactive matrix with improved angiogenic and anti-inflammatory capacities (SNF-AC-Mg). Mg2+ and AC distributed homogeneously inside the silk nanofiber hydrogels without compromising the mechanical performance. Both Mg2+ and AC released slowly to continuously tune both angiogenic and inflammatory behaviors. The hydrogels exhibited good biocompatibility, inflammation inhibition, and pro-angiogenic properties in vitro, suggesting the synergistic bioactivity of AC and Mg2+. In vivo analysis revealed that the synergistic action of AC and Mg2+ resulted in better M2-type polarization of macrophages and angiogenesis during the inflammatory phase, while effectively achieving the inhibition of excessive accumulation of collagen and scar formation during the remodeling phases. The quicker scarless regeneration of the defects treated with SNF-AC-Mg implies the priority of SNFs in designing bioactive niches with complicated cues, which will favor the functional recovery of different tissues in the future.
Collapse
Affiliation(s)
- Gongwen Yang
- State Key Laboratory of Radiation Medicine and Radiation Protection, Institutes for Translational Medicine, Soochow University, Suzhou 215123, People's Republic of China.
| | - Lutong Liu
- Beijing Allgens Medical Science and Technology Co., Ltd., Beijing 100176, People's Republic of China
| | - Liying Xiao
- State Key Laboratory of Radiation Medicine and Radiation Protection, Institutes for Translational Medicine, Soochow University, Suzhou 215123, People's Republic of China.
- National Engineering Laboratory for Modern Silk & Collaborative Innovation Center of Suzhou Nano Science and Technology, Soochow University, Suzhou 215123, People's Republic of China
| | - Shiyu Ke
- National Engineering Laboratory for Modern Silk & Collaborative Innovation Center of Suzhou Nano Science and Technology, Soochow University, Suzhou 215123, People's Republic of China
| | - Huaxiang Yang
- National Engineering Laboratory for Modern Silk & Collaborative Innovation Center of Suzhou Nano Science and Technology, Soochow University, Suzhou 215123, People's Republic of China
| | - Qiang Lu
- State Key Laboratory of Radiation Medicine and Radiation Protection, Institutes for Translational Medicine, Soochow University, Suzhou 215123, People's Republic of China.
| |
Collapse
|
22
|
Xu J, Zhang C, Yan Z, Fan C, Yuan S, Wang J, Zhu Y, Luo L, Shi K, Deng J. Dental Pulp Stem Cell Lysate-Based Hydrogel Improves Diabetic Wound Healing via the Regulation of Anti-Inflammatory Macrophages and Keratinocytes. ACS APPLIED BIO MATERIALS 2024; 7:7684-7699. [PMID: 39503733 DOI: 10.1021/acsabm.4c01157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2024]
Abstract
The prolonged existence of chronic wounds heightens the risk of patients experiencing chronic pain, necrosis, and amputation. Dental pulp stem cells (DPSCs) have garnered attention due to their potential immunomodulatory and tissue repair regenerative effects in the management of chronic wounds. However, stem-cell-based therapy faces challenges such as malignant differentiation, immune rejection, and long-term effectiveness. To overcome these challenges, we proposed a chronic wound therapy using a hydrogel derived from human-originated dental pulp stem cell lysate (DPSCL). Our data indicate that, with the degradation of the dental pulp stem cell lysate-based hydrogel (DPSCLH), the slowly released cell lysates recruit anti-inflammatory M2 macrophages and promote the proliferation, migration, and keratinization of HacaT cells. In addition, in vivo studies revealed that DPSCLH avoids immune rejection reactions and induces a long-term accumulation of endogenous M2 macrophages. In a mouse model of diabetic wounds, DPSCLH effectively modulates the inflammatory microenvironment around diabetic wounds, promotes the formation of the stratum corneum, and facilitates the healing of wounds, thus holding tremendous potential for the treatment of diabetic wounds.
Collapse
Affiliation(s)
- Jianghua Xu
- Joint Centre of Translational Medicine, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325000, China
- Joint Centre of Translational Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
- Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325000, China
| | - Changhuan Zhang
- First People's Hospital of Linping District, Hangzhou, Hangzhou, Zhejiang 311100, China
| | - Zhuo Yan
- Joint Centre of Translational Medicine, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325000, China
- Joint Centre of Translational Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
- Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325000, China
| | - Chen Fan
- Joint Centre of Translational Medicine, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325000, China
- Joint Centre of Translational Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
- Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325000, China
| | - Shanshan Yuan
- Joint Centre of Translational Medicine, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325000, China
- Joint Centre of Translational Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
- Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325000, China
| | - Jilong Wang
- Joint Centre of Translational Medicine, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325000, China
- Joint Centre of Translational Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
- Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325000, China
| | - Yuting Zhu
- Joint Centre of Translational Medicine, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325000, China
- Joint Centre of Translational Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
- Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325000, China
| | - Lihua Luo
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Keqing Shi
- Joint Centre of Translational Medicine, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325000, China
- Joint Centre of Translational Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Junjie Deng
- Joint Centre of Translational Medicine, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325000, China
- Joint Centre of Translational Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
- Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325000, China
| |
Collapse
|
23
|
Trotzier C, Bellanger C, Abdessadeq H, Delannoy P, Mojallal A, Auxenfans C. Deciphering influence of donor age on adipose-derived stem cells: in vitro paracrine function and angiogenic potential. Sci Rep 2024; 14:27589. [PMID: 39528480 PMCID: PMC11555058 DOI: 10.1038/s41598-024-73875-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 09/23/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND As fat grafting is commonly used as a filler, Adipose-derived stem/stromal cells (ASC) have been reported to be key player in retention rate. Paracrine and differentiation potential of those cells confer them strong pro-angiogenic capacities. However, a full characterization of the influence of aging on ASC has not been reported yet. Here we've investigated the effect of age on paracrine function, stemness and angiogenic potential of ASC. METHODS ASC were extracted from young and old adult donors. We assessed stromal vascular fraction cell populations repartition, ASC stemness potential, capability to differentiate into mesenchymal lineages as well as their secretome. Angiogenic potential was assessed using a sprouting assay, an indirect co-culture of ASC and dermal microvascular endothelial cells (EC). Total vascular sprout length was measured, and co-culture soluble factors were quantified. Pro-angiogenic factors alone or in combination as well as ASC-conditioned medium (CM) were added to EC to assess sprouting induction. RESULTS Decrease of endothelial cells yield and percentage is observed in cells extracted from adipose tissue of older patients, whereas ASC percentage increased with age. Clonogenic potential of ASC is stable with age. ASC can differentiate into adipocytes, chondrocytes and osteoblasts, and aging does not alter this potential. Among the 25 analytes quantified, high levels of pro-angiogenic factors were found, but none is significantly modulated with age. ASC induce a significantly longer vascular sprouts compared to fibroblasts, and no difference was found between young and old ASC donors on that parameter. Higher concentrations of FGF-2, G-CSF, HGF and IL-8, and lower concentrations of VEGF-C were quantified in EC/ASC co-cultures compared to EC/fibroblasts co-cultures. EC/ASC from young donors secrete higher levels of VEGF-A compared to old ones. Neither soluble factor nor CM without cells are able to induce organized sprouts, highlighting the requirement of cell communication for sprouting. CM produced by ASC supporting development of long vascular sprouts promote sprouting in co-cultures that establish shorter sprouts. CONCLUSION Our results show cells from young and old donors exhibit no difference in all assessed parameters, suggesting all patients could be included in clinical applications. We emphasized the leading role of ASC in angiogenesis, without impairment with age, where secretome is a key but not sufficient actor.
Collapse
Affiliation(s)
- Chloe Trotzier
- Advanced Research, L'Oréal Research and Innovation, 1, Av. Eugene Schueller, 93600, Aulnay sous Bois, France.
| | - Clement Bellanger
- Advanced Research, L'Oréal Research and Innovation, 1, Av. Eugene Schueller, 93600, Aulnay sous Bois, France
| | - Hakima Abdessadeq
- Advanced Research, L'Oréal Research and Innovation, 1, Av. Eugene Schueller, 93600, Aulnay sous Bois, France
| | - Philippe Delannoy
- Advanced Research, L'Oréal Research and Innovation, 1, Av. Eugene Schueller, 93600, Aulnay sous Bois, France
| | - Ali Mojallal
- Department of Plastic, Reconstructive and Aesthetic Surgery, La Croix Rousse Hospital, Bernard Lyon 1 University, Lyon, France
| | - Celine Auxenfans
- Banque de Tissus et de Cellules des Hospices Civils de Lyon, Edouard Herriot Hospital, Lyon, France
| |
Collapse
|
24
|
Li C, Ren S, Yan C, Wang C, Jiang T, Kang Y, Chen J, Xiong H, Guo J, Jiang G, Liu S, Nie P, Chen Z. HES1 revitalizes the functionality of aged adipose-derived stem cells by inhibiting the transcription of STAT1. Stem Cell Res Ther 2024; 15:399. [PMID: 39501364 PMCID: PMC11539794 DOI: 10.1186/s13287-024-04002-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Accepted: 10/16/2024] [Indexed: 11/08/2024] Open
Abstract
BACKGROUND The effectiveness of adipose-derived stem cells (ADSCs) in therapy diminishes with age. It has been reported that transcription factors (TFs) play a crucial role in the aging and functionality of stem cells. Nevertheless, there is limited understanding regarding the involvement of TFs in the aging mechanism of ADSCs. METHODS RNA sequencing (RNA-seq) was utilized to discern the differentially expressed genes in ADSCs obtained from donors of varying ages. TFs exhibiting significant variations across age groups were identified and subsequently validated. ADSCs were manipulated to exhibit either enhanced expression or reduced levels of HES1 and STAT1 via lentivirus transfection and small interfering RNA (siRNA) techniques. The impact of these genetic alterations on ADSCs' proliferation, migration, and cellular senescence was assessed using EdU, transwell, and senescence-activated β-galactosidase (SA-β-gal) staining assays. The DNA sequences bound by HES1 were investigated through the CUT & Tag assay. Lastly, the therapeutic efficacy of aged ADSCs with HES1 overexpression was evaluated in skin injury model of male Sprague-Dawley rats. RESULTS 678 genes showed differential expression between ADSCs obtained from young and old donors (Y-ADSCs and O-ADSCs), with 47 of these genes being TFs. Notably, the expression of the TF hairy and enhancer of split 1 (HES1) was notably reduced in ADSCs from old donors. Introducing HES1 overexpression in aged ADSCs resulted in improved cellular function and the suppression of cellular senescence, while reducing HES1 levels in young ADSCs had the opposite effect. Mechanistically, HES1 was found to interact with the promoter region of another TF, signal transducer and activator of transcription 1 (STAT1), to inhibit its transcription. Knocking down STAT1 could fully reverse the negative effects caused by decreased HES1 in ADSCs, leading to a reduction in the secretion of pro-inflammatory cytokines such as TNF-α, IL-6, and IL-8. Ultimately, restoring HES1 expression in aged ADSCs demonstrated enhanced therapeutic potential in promoting skin wound healing. CONCLUSION HES1 acts as an inhibitor of cellular senescence in the aging progression of ADSCs through the modulation of STAT1 expression, suggesting a promising avenue for rejuvenating senescent ADSCs and improving wound healing.
Collapse
Affiliation(s)
- Chengcheng Li
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, 430022, China
| | - Sen Ren
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Chengqi Yan
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, 430022, China
| | - Cheng Wang
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, 430022, China
| | - Tao Jiang
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, 430022, China
| | - Yu Kang
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, 430022, China
| | - Jing Chen
- Department of Dermatology, Wuhan No.1 Hospital, Wuhan, Hubei, 430000, China
| | - Hewei Xiong
- Department of Emergency Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Jiahe Guo
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, 430022, China
| | - Guoyong Jiang
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, 430022, China
| | - Shuoyuan Liu
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, 430022, China
| | - Pengjuan Nie
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, 430022, China
| | - Zhenbing Chen
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, 430022, China.
| |
Collapse
|
25
|
Yang Z, Yang M, Rui S, Hao W, Wu X, Guo L, Armstrong DG, Yang C, Deng W. Exosome-based cell therapy for diabetic foot ulcers: Present and prospect. Heliyon 2024; 10:e39251. [PMID: 39498056 PMCID: PMC11532254 DOI: 10.1016/j.heliyon.2024.e39251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 09/17/2024] [Accepted: 10/10/2024] [Indexed: 10/30/2024] Open
Abstract
Diabetic foot ulcers (DFUs) represent a serious complication of diabetes with high incidence, requiring intensive treatment, prolonged hospitalization, and high costs. It poses a severe threat to the patient's life, resulting in substantial burdens on patient and healthcare system. However, the therapy of DFUs remains challenging. Therefore, exploring cell-free therapies for DFUs is both critical and urgent. Exosomes, as crucial mediators of intercellular communication, have been demonstrated potentially effective in anti-inflammation, angiogenesis, cell proliferation and migration, and collagen deposition. These functions have been proven beneficial in all stages of diabetic wound healing. This review aims to summarize the role and mechanisms of exosomes from diverse cellular sources in diabetic wound healing research. In addition, we elaborate on the challenges for clinical application, discuss the advantages of membrane vesicles as exosome mimics in wound healing, and present the therapeutic potential of exosomes and their mimetic vesicles for future clinical applications.
Collapse
Affiliation(s)
- Zhou Yang
- Department of Endocrinology and Metabolism, School of Medicine, Chongqing University Central Hospital, Chongqing Emergency Medical Center, Chongqing, 400014, China
| | - Mengling Yang
- Department of Endocrinology and Metabolism, School of Medicine, Chongqing University Central Hospital, Chongqing Emergency Medical Center, Chongqing, 400014, China
| | - Shunli Rui
- Department of Endocrinology and Metabolism, School of Medicine, Chongqing University Central Hospital, Chongqing Emergency Medical Center, Chongqing, 400014, China
| | - Wei Hao
- Department of Endocrinology and Metabolism, School of Medicine, Chongqing University Central Hospital, Chongqing Emergency Medical Center, Chongqing, 400014, China
| | - Xiaohua Wu
- Department of Endocrinology and Metabolism, School of Medicine, Chongqing University Central Hospital, Chongqing Emergency Medical Center, Chongqing, 400014, China
| | - Lian Guo
- Department of Endocrinology, School of Medicine, Chongqing University Three Gorges Central Hospital, Chongqing, 404000, China
| | - David G. Armstrong
- Department of Surgery, Keck School of Medicine of University of Southern California, Los Angeles, CA, 90033, USA
| | - Cheng Yang
- Department of Endocrinology and Metabolism, School of Medicine, Chongqing University Central Hospital, Chongqing Emergency Medical Center, Chongqing, 400014, China
| | - Wuquan Deng
- Department of Endocrinology and Metabolism, School of Medicine, Chongqing University Central Hospital, Chongqing Emergency Medical Center, Chongqing, 400014, China
| |
Collapse
|
26
|
Zhang K, Xiao D, Li F, Song G, Huang G, Wang Y, Liu H. Combination therapy of placenta-derived mesenchymal stem cells and artificial dermal scaffold promotes full-thickness skin defects vascularization in rat animal model. Adv Med Sci 2024; 70:8-16. [PMID: 39424001 DOI: 10.1016/j.advms.2024.10.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 07/14/2024] [Accepted: 10/14/2024] [Indexed: 10/21/2024]
Abstract
PURPOSE Recently, placenta-derived mesenchymal stem cells (PMSCs) have garnered considerable attention in tissue repair and regeneration. The present study was conducted to evaluate the effect of PMSCs on artificial dermal scaffold (ADS) angiogenesis and their combination therapy on wound closure. MATERIAL AND METHODS Herein, the growth and survival of PMSCs in ADS were explored. CCK8, scratch wound, and tubule formation assays were employed to investigate the effects of ADS conditioned medium (CM) and ADS-PMSCs CM on human umbilical vein endothelial cells (HUVECs). The effect of ADS-PMSCs on full-thickness skin defects healing was evaluated based on a rat model. Wound healing progresses was meticulously investigated through hematoxylin and eosin (HE), Masson's trichrome, and immunohistochemical staining analyses. RESULTS In vitro cell culture results demonstrated the proliferation of PMSCs in ADS. The ADS-PMSCs CM notably stimulated the proliferation, migration, and tube formation of HUVECs compared to the ADS CM group. In the rat full-thickness skin defect model, the ADS-PMSCs treatment significantly accelerated the vascularization area of ADS after 2 weeks. Besides, HE and Masson's trichrome staining results indicated that ADS-PMSCs treatment significantly enhanced fibroblast proliferation and collagen fiber 2 weeks after surgical procedure. Compared to the ADS group, collagen fiber arrangement was thicker in the ADS-PMSCs group. Immunohistochemical staining reinforced this finding, illustrating a substantial increase in CD31 expression within the ADS-PMSCs group. CONCLUSIONS The results suggest that the combination of ADS with PMSCs accelerates ADS vascularization by fostering granulation tissue development and boosting the formation of new blood vessels.
Collapse
Affiliation(s)
- Kun Zhang
- Cell Therapy Center, Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Dongjie Xiao
- Cell Therapy Center, Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Fang Li
- Cell Therapy Center, Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Guodong Song
- Department of Burns and Plastic Surgery, Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Guobao Huang
- Department of Burns and Plastic Surgery, Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Yunshan Wang
- Cell Therapy Center, Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Hua Liu
- Cell Therapy Center, Central Hospital Affiliated to Shandong First Medical University, Jinan, China.
| |
Collapse
|
27
|
Matsuoka T, Dan K, Takanashi K, Ogino A. Early Effects of Porcine Placental Extracts and Stem Cell-Derived Exosomes on Aging Stress in Skin Cells. J Funct Biomater 2024; 15:306. [PMID: 39452604 PMCID: PMC11509013 DOI: 10.3390/jfb15100306] [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: 09/24/2024] [Revised: 10/07/2024] [Accepted: 10/12/2024] [Indexed: 10/26/2024] Open
Abstract
The initial efficacy of placental extracts (Pla-Exts) and human mesenchymal stem-cell-derived exosomes (hMSC-Exo) against aging-induced stress in human dermal fibroblasts (HDFs) was examined. The effect of Pla-Ext alone, hMSC-Exo alone, the combined effect of Pla-Ext and hMSC-Exo, and the effect of hMSC-Exo (Pla/MSC-Exo) recovered from cultures with Pla-Ext added to hMSC were verified using collagen, elastin, and hyaluronic acid synthase mRNA levels for each effect. Cells were subjected to photoaging (UV radiation), glycation (glycation end-product stimulation), and oxidation (H2O2 stimulation) as HDF stressors. Pla-Ext did not significantly affect normal skin fibroblasts with respect to intracellular parameters; however, a pro-proliferative effect was observed. Pla-Ext induced resistance to several stresses in skin fibroblasts (UV irradiation, glycation stimulation, H2O2 stimulation) and inhibited reactive oxygen species accumulation following H2O2 stimulation. Although the effects of hMSC-Exo alone or the combination of hMSC-Exo and Pla-Ext are unknown, pretreated hMSC-Exo stimulated with Pla-Ext showed changes that conferred resistance to aging stress. This suggests that Pla-Ext supplementation may cause some changes in the surface molecules or hMSC-Exo content (e.g., microRNA). In skin cells, the direct action of Pla-Ext and exosomes secreted from cultured hMSCs pretreated with Pla-Ext (Pla/MSC-Exo) also conferred resistance to early aging stress.
Collapse
Affiliation(s)
| | - Katsuaki Dan
- Department of Pathophysiology, Yokohama University of Pharmacy, Yokohama-shi 245-0066, Kanagawa, Japan; (K.D.); (K.T.)
- Division of Research and Development, Research Organization of Biological Activity, Shibuya-ku 150-0001, Tokyo, Japan
| | - Keita Takanashi
- Department of Pathophysiology, Yokohama University of Pharmacy, Yokohama-shi 245-0066, Kanagawa, Japan; (K.D.); (K.T.)
| | - Akihiro Ogino
- Department of Plastic and Reconstructive Surgery, Toho University Omori Medical Center, Ota-ku 143-8541, Tokyo, Japan;
| |
Collapse
|
28
|
Kong W, Bao Y, Li W, Guan D, Yin Y, Xiao Y, Zhu S, Sun Y, Xia Z. Collaborative Enhancement of Diabetic Wound Healing and Skin Regeneration by Recombinant Human Collagen Hydrogel and hADSCs. Adv Healthc Mater 2024:e2401012. [PMID: 39388509 DOI: 10.1002/adhm.202401012] [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: 03/18/2024] [Revised: 09/09/2024] [Indexed: 10/12/2024]
Abstract
Stem cell-based therapies hold significant promise for chronic wound healing and skin appendages regeneration, but challenges such as limited stem cell lifespan and poor biocompatibility of delivery systems hinder clinical application. In this study, an in situ delivery system for human adipose-derived stem cells is developed (hADSCs) to enhance diabetic wound healing. The system utilizes a photo-crosslinking recombinant human type III collagen (rHCIII) hydrogel to encapsulate hADSCs, termed the hADSCs@rHCIII hydrogel. This hydrogel undergoes local crosslinking at the wound site, establishing a sturdy 3D niche suitable for stem cell function. Consequently, the encapsulated hADSCs exhibit strong attachment and spreading within the hydrogels, maintaining their proliferation, metabolic activity, and viability for up to three weeks in vitro. Importantly, in vivo studies demonstrate that the hADSCs@rHCIII hydrogel achieves significant in situ delivery of stem cells, prolonging their retention within the wound. This ultimately enhances their immunomodulatory capabilities, promotes neovascularization and granulation tissue formation, facilitates matrix remodeling, and accelerates healing in a diabetic mouse wound model. Collectively, these findings highlight the potential of the conveniently-prepared and user-friendly hADSCs@rHCIII hydrogel as a promising therapeutic approach for diabetic wound treatment and in situ skin regeneration.
Collapse
Affiliation(s)
- Weishi Kong
- Department of Burn Surgery, the First Affiliated Hospital, Naval Medical University, Shanghai, 200433, P. R. China
- Research Unit of key techniques for treatment of burns and combined burns and trauma injury, Chinese Academy of Medical Sciences, Shanghai, 200433, P. R. China
| | - Yulu Bao
- Department of Burn Surgery, the First Affiliated Hospital, Naval Medical University, Shanghai, 200433, P. R. China
- Research Unit of key techniques for treatment of burns and combined burns and trauma injury, Chinese Academy of Medical Sciences, Shanghai, 200433, P. R. China
| | - Wei Li
- Department of Burn Surgery, the First Affiliated Hospital, Naval Medical University, Shanghai, 200433, P. R. China
- Research Unit of key techniques for treatment of burns and combined burns and trauma injury, Chinese Academy of Medical Sciences, Shanghai, 200433, P. R. China
| | - Dingding Guan
- Department of Burn Surgery, the First Affiliated Hospital, Naval Medical University, Shanghai, 200433, P. R. China
- Research Unit of key techniques for treatment of burns and combined burns and trauma injury, Chinese Academy of Medical Sciences, Shanghai, 200433, P. R. China
| | - Yating Yin
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, P. R. China
- Department of Burn and Plastic Surgery, Seventh People's Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200137, P. R. China
| | - Yongqiang Xiao
- Department of Burn Surgery, the First Affiliated Hospital, Naval Medical University, Shanghai, 200433, P. R. China
- Research Unit of key techniques for treatment of burns and combined burns and trauma injury, Chinese Academy of Medical Sciences, Shanghai, 200433, P. R. China
- ENT Institute, Department of Facial Plastic and Reconstructive Surgery, Eye & ENT Hospital, Fudan University, Shanghai, 200031, P. R. China
| | - Shihui Zhu
- Department of Burn Surgery, the First Affiliated Hospital, Naval Medical University, Shanghai, 200433, P. R. China
- Research Unit of key techniques for treatment of burns and combined burns and trauma injury, Chinese Academy of Medical Sciences, Shanghai, 200433, P. R. China
- Department of Burns and Plastic Surgery, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, P. R. China
| | - Yu Sun
- Department of Burn Surgery, the First Affiliated Hospital, Naval Medical University, Shanghai, 200433, P. R. China
- Research Unit of key techniques for treatment of burns and combined burns and trauma injury, Chinese Academy of Medical Sciences, Shanghai, 200433, P. R. China
| | - Zhaofan Xia
- Department of Burn Surgery, the First Affiliated Hospital, Naval Medical University, Shanghai, 200433, P. R. China
- Research Unit of key techniques for treatment of burns and combined burns and trauma injury, Chinese Academy of Medical Sciences, Shanghai, 200433, P. R. China
| |
Collapse
|
29
|
Xue C, Chen L, Wang N, Chen H, Xu W, Xi Z, Sun Q, Kang R, Xie L, Liu X. Stimuli-responsive hydrogels for bone tissue engineering. BIOMATERIALS TRANSLATIONAL 2024; 5:257-273. [PMID: 39734705 PMCID: PMC11681187 DOI: 10.12336/biomatertransl.2024.03.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 07/22/2024] [Accepted: 08/30/2024] [Indexed: 12/31/2024]
Abstract
The treatment of bone defects remains a great clinical challenge. With the development of science and technology, bone tissue engineering technology has emerged, which can mimic the structure and function of natural bone tissues and create solutions for repairing or replacing human bone tissues based on biocompatible materials, cells and bioactive factors. Hydrogels are favoured by researchers due to their high water content, degradability and good biocompatibility. This paper describes the hydrogel sources, roles and applications. According to the different types of stimuli, hydrogels are classified into three categories: physical, chemical and biochemical responses, and the applications of different stimuli-responsive hydrogels in bone tissue engineering are summarised. Stimuli-responsive hydrogels can form a semi-solid with good adhesion based on different physiological environments, which can carry a variety of bone-enhancing bioactive factors, drugs and cells, and have a long retention time in the local area, which is conducive to a long period of controlled release; they can also form a scaffold for constructing tissue repair, which can jointly promote the repair of bone injury sites. However, it also has many defects, such as poor biocompatibility, immunogenicity and mechanical stability. Further studies are still needed in the future to facilitate its clinical translation.
Collapse
Affiliation(s)
- Congyang Xue
- Department of Orthopaedics, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province, China
- The Third Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province, China
| | - Liping Chen
- Department of Orthopaedics, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province, China
- Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, Jiangsu Province, China
| | - Nan Wang
- Department of Orthopaedics, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province, China
| | - Heng Chen
- Department of Orthopaedics, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province, China
| | - Wenqiang Xu
- Department of Orthopaedics, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province, China
- Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, Jiangsu Province, China
| | - Zhipeng Xi
- Department of Orthopaedics, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province, China
- Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, Jiangsu Province, China
| | - Qing Sun
- Laboratory of Gene Therapy, Faculty of Veterinary Medicine, Ghent University, Ghent, Belgium
| | - Ran Kang
- Department of Orthopaedics, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province, China
- Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, Jiangsu Province, China
| | - Lin Xie
- Department of Orthopaedics, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province, China
- Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, Jiangsu Province, China
| | - Xin Liu
- Department of Orthopaedics, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province, China
- The Third Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province, China
- Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, Jiangsu Province, China
| |
Collapse
|
30
|
Liu C, Khairullina L, Qin Y, Zhang Y, Xiao Z. Adipose stem cell exosomes promote mitochondrial autophagy through the PI3K/AKT/mTOR pathway to alleviate keloids. Stem Cell Res Ther 2024; 15:305. [PMID: 39278919 PMCID: PMC11403874 DOI: 10.1186/s13287-024-03928-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 09/05/2024] [Indexed: 09/18/2024] Open
Abstract
BACKGROUND Fibrosis with unrelieved chronic inflammation is an important pathological change in keloids. Mitochondrial autophagy plays a crucial role in reducing inflammation and inhibiting fibrosis. Adipose stem cell-derived exosomes, a product of adipose stem cell paracrine secretion, have pharmacological effects, such as anti-inflammatory and antiapoptotic effects, and mediate autophagy. Therefore, this study aims to investigate the function and mechanism of adipose stem cell exosomes in the treatment of keloids. METHOD We isolated adipose stem cell exosomes under normoxic and hypoxic condition to detect their effects on keloid fibroblast proliferation, migration, and collagen synthesis. Meanwhile, 740YPDGFR (PI3K/AKT activator) was applied to detect the changes in autophagic flow levels and mitochondrial morphology and function in keloid fibroblasts. We constructed a human keloid mouse model by transplanting human keloid tissues into six-week-old (20-22 g; female) BALB/c nude mice, meanwhile, we applied adipose stem cell exosomes to treat the mouse model and observed the retention and effect of ADSC exosomes in vivo. RESULTS ADSC exosomes can inhibit the PI3K/AKT/mTOR signaling pathway. The exosomes of ADSCs decreased the inflammatory level of KFs, enhanced the interaction between P62 and LC3, and restored the mitochondrial membrane potential. In the human keloid mouse model, ADSC exosomes can exist stably, promote mitochondrial autophagy in keloid tissue, improve mitochondrial morphology, reduce inflammatory reaction and fibrosis. Meanwhile, At the same time, the exosomes derived from hypoxic adipose stem cells have played a more effective role in both in vitro and in vivo experiments. CONCLUSIONS Adipose stem cell exosomes inhibited the PI3K/AKT/mTOR pathway, activated mitochondrial autophagy, and alleviated keloid scars.
Collapse
Affiliation(s)
- Chang Liu
- Department of Plastic and Aesthetic Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150081, China
| | - Liliia Khairullina
- Department of Plastic and Aesthetic Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150081, China
| | - Youyou Qin
- Department of Plastic and Aesthetic Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150081, China
| | - Yingbo Zhang
- Department of Plastic and Aesthetic Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150081, China
| | - Zhibo Xiao
- Department of Plastic and Aesthetic Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150081, China.
| |
Collapse
|
31
|
Gu Y, Mu Z, Chen Y, Wu C, Shi J, Bai N. Therapeutic potential of ADSCs in diabetic wounds: a proteomics-based approach. Front Cell Dev Biol 2024; 12:1468220. [PMID: 39345337 PMCID: PMC11427884 DOI: 10.3389/fcell.2024.1468220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Accepted: 08/26/2024] [Indexed: 10/01/2024] Open
Abstract
Background Diabetes mellitus (DM), a chronic metabolic disease characterized by elevated blood sugar, leads to delayed or non-healing wounds, increasing amputation risks, and placing a significant burden on patients and society. While extensive research has been conducted on adipose-derived stem cells (ADSCs) for promoting wound healing, there is a scarcity of studies focusing on diabetic wounds, particularly those employing proteomics and bioinformatics approaches. Objective This study aimed to investigate the mechanisms by which ADSCs promote diabetic wound healing using proteomics and bioinformatics techniques. Methods Healthy rat fat tissue was used to isolate ADSCs. A T2DM rat model with back wounds was established. The experimental group received ADSC injections around the wound, while the control group received PBS injections. Wound healing rates were documented and photographed on days 0, 3, 7, 10, and 14. On day 7, wound tissues were excised for HE and Masson's staining. Additionally, on day 7, tissues were analyzed for protein quantification using 4D-DIA, with subsequent GO and KEGG analyses for differentially expressed proteins (DEPs) and protein-protein interaction (PPI) network analysis using STRING database (String v11.5). Finally, Western blot experiments were performed on day 7 wounds to verify target proteins. Results and Conclusions In all measured days postoperatively, the wound healing rate was significantly higher in the ADSC group than in the PBS group (day 7: p < 0.001, day 10: p = 0.001, day 14: p < 0.01), except on day 3 (p > 0.05). Proteomic analysis identified 474 differentially expressed proteins, with 224 key proteins after PPI analysis (78 upregulated and 146 downregulated in the ADSC group). The main cellular locations of these proteins were "cellular anatomical entity" and "protein-containing complex", while the biological processes were "cellular processes" and "biological regulation". The primary molecular functions were "binding" and "catalytic activity", with GO enrichment focused on "Wnt-protein binding", "neural development", and "collagen-containing extracellular matrix". Further analysis of PPI network nodes using LASSO regression identified Thy1 and Wls proteins, significantly upregulated in the ADSC group, as potentially crucial targets for ADSC application in diabetic wound treatment.
Collapse
Affiliation(s)
- Yuan Gu
- School of Clinical Medicine, Shandong Second Medical University, Weifang, China
| | - Zelan Mu
- School of Clinical Medicine, Shandong Second Medical University, Weifang, China
| | - Yuanzheng Chen
- Department of Burns and Plastic Surgery, Emergency General Hospital, Beijing, China
| | - Can Wu
- Medical Cosmetic Plastic Surgery, Linyi People′s Hospital, Linyi, China
| | - Jie Shi
- Plastic and Cosmetic Surgery, People′s Hospital of Liaoning Province, Shenyang, China
| | - Nan Bai
- Medical Cosmetic Plastic Surgery, Linyi People′s Hospital, Linyi, China
| |
Collapse
|
32
|
Ma Z, Ding J, Liu J, Zhao Z, Long Q, Zhang T, Huang J. Effect of baicalin combined with adipose stem cells on promoting angiogenesis and improving wound healing in diabetes. Panminerva Med 2024; 66:324-326. [PMID: 38990211 DOI: 10.23736/s0031-0808.24.05068-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/12/2024]
Affiliation(s)
- Zhibing Ma
- Department of Plastic Surgery, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Jin Ding
- Department of Pathology, Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, China
| | - Jia Liu
- Department of Plastic Surgery, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Zhijie Zhao
- Department of Plastic Surgery, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Qiuzi Long
- Department of Plastic Surgery, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Tianqi Zhang
- Department of Plastic Surgery, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Jinlong Huang
- Department of Plastic Surgery, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China -
| |
Collapse
|
33
|
Tang W, Du X, Wu Z, Nie Z, Yu C, Gao Y. circ-Erbb2ip from adipose-derived mesenchymal stem cell-derived exosomes promotes wound healing in diabetic mice by inducing the miR-670-5p/Nrf1 axis. Cell Signal 2024; 121:111245. [PMID: 38849105 DOI: 10.1016/j.cellsig.2024.111245] [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: 10/10/2023] [Revised: 03/16/2024] [Accepted: 06/04/2024] [Indexed: 06/09/2024]
Abstract
BACKGROUND To investigate the mechanism of exosomes (Exo) secretion by hypoxic pretreated adipose-derived mesenchymal stem cells (ADSCs) promoting skin wound healing in diabetic (DM) mice. METHODS High-throughput sequencing was used to investigate abnormal expression of circRNA in hypoxic pretreatment ADSCs exosome (HExo) and ADSCs exosome (Exo). Bioinformatics analysis and luciferase reporting analysis were used to clarify the interacted relationship among circRNA, miRNA and mRNA. EPCs cells were employ to analysis the ROS, inflammatory cytokines expression, angiogenic differentiation function under hypoxic condition by using immunofluorescence, ELISA detection and tube forming experiment. DM ulceration mice model were constructed and the therapeutic effect of Exo were detected using immunohistochemistry, immunofluorescence. RESULTS The result show that HExo have more treatment effect than Exo in promotes cutaneous wound healing of DM mice. High-throughput sequencing found that circ-Erbb2ip play a role in HExo mediated tissues repair. Downregulation circ-Erbb2ip decreased the therapeutic effect of HExo to wound healing in diabetic mice. Bioinformatics analysis and luciferase reporting analysis confirmed that both miR-670-5p and Nrf1 were downstream targets of circ-Erbb2ip. Downregulation of Nrf1 or overexpression of miR-670-5p reversed the protective effect of circ-Erbb2ip to EPCs after exposure to high glucose microenvironment. Upregulation circ-Erbb2ip increased the therapeutic effect of Exo to wound healing in diabetic mice by increased angiogenesis and decreased ROS, inflammatory cytokines expression. CONCLUSION In conclusion, ADSC-Exos containing circ-Erbb2ip promotes wound healing by targeting miR-670-5p/Nrf1 pathway, and their effects in promoting soft tissue wound healing warrant further study.
Collapse
Affiliation(s)
- Wenbo Tang
- The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong 510632, China; The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui 233000, China
| | - Xiaoying Du
- The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong 510632, China; The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui 233000, China
| | - Zifu Wu
- The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui 233000, China
| | - Zhonglin Nie
- The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui 233000, China
| | - Chaowen Yu
- The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui 233000, China.
| | - Yong Gao
- The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong 510632, China; The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui 233000, China.
| |
Collapse
|
34
|
Ou H, Yang Q, Zhang Y, Tang X, Xiao M, Li S, Lei L, Xie Z. The role of cells and their derivatives in otorhinolaryngologic diseases treatment. Life Sci 2024; 352:122898. [PMID: 38997061 DOI: 10.1016/j.lfs.2024.122898] [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: 04/13/2024] [Revised: 06/23/2024] [Accepted: 07/05/2024] [Indexed: 07/14/2024]
Abstract
Otolaryngology is an important specialty in the field of surgery that deals with the diagnosis and treatment of the ear, nose, throat, trachea, as well as related anatomical structures. Various otolaryngological disorders are difficult to treat using established pharmacological and surgical approaches. The advent of molecular and cellular therapies led to further progress in this respect. This article reviews the therapeutic strategies of using stem cells, immune cells, and chondrocytes in otorhinolaryngology. As the most widely recognized cell derivatives, exosomes were also systematically reviewed for their therapeutic potential in head and neck cancer, otitis media, and allergic rhinitis. Finally, we summarize the limitations of stem cells, chondrocytes, and exosomes, as well as possible solutions, and provide an outlook on the future direction of cell- and derivative-based therapies in otorhinolaryngology, to offer a theoretical foundation for the clinical translation of this therapeutic modality.
Collapse
Affiliation(s)
- Haibo Ou
- Department of Otorhinolaryngology Head and Neck Surgery, the Second Xiangya Hospital, Central South University, Changsha 410011, Hunan, China
| | - Qian Yang
- Department of Otorhinolaryngology Head and Neck Surgery, the Second Xiangya Hospital, Central South University, Changsha 410011, Hunan, China
| | - Yuming Zhang
- Department of Otorhinolaryngology Head and Neck Surgery, the Second Xiangya Hospital, Central South University, Changsha 410011, Hunan, China
| | - Xiaojun Tang
- Department of Otorhinolaryngology Head and Neck Surgery, the Second Xiangya Hospital, Central South University, Changsha 410011, Hunan, China
| | - Minna Xiao
- Department of Otorhinolaryngology Head and Neck Surgery, the Second Xiangya Hospital, Central South University, Changsha 410011, Hunan, China
| | - Shisheng Li
- Department of Otorhinolaryngology Head and Neck Surgery, the Second Xiangya Hospital, Central South University, Changsha 410011, Hunan, China
| | - Lanjie Lei
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Institute of Translational Medicine, Zhejiang Shuren University, Hangzhou 310015, Zhejiang, China.
| | - Zuozhong Xie
- Department of Otorhinolaryngology Head and Neck Surgery, the Second Xiangya Hospital, Central South University, Changsha 410011, Hunan, China.
| |
Collapse
|
35
|
Shah S, Lucke-Wold B. Image-Guided Mesenchymal Stem Cell Sodium Iodide Symporter (NIS) Radionuclide Therapy for Glioblastoma. Cancers (Basel) 2024; 16:2892. [PMID: 39199662 PMCID: PMC11352884 DOI: 10.3390/cancers16162892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 08/12/2024] [Accepted: 08/20/2024] [Indexed: 09/01/2024] Open
Abstract
BACKGROUND Glioblastoma (GBM) is a highly aggressive, invasive, and growth factor-independent grade IV glioma. Survival following the diagnosis is generally poor, with a median survival of approximately 15 months, and it is considered the most aggressive and lethal central nervous system tumor. Conventional treatments based on surgery, chemotherapy, and radiation therapy only delay progression, and death is inevitable. Malignant glioma cells are resistant to traditional therapies, potentially due to a subpopulation of glioma stem cells that are invasive and capable of rapid regrowth. METHODS This is a literature review. The systematic retrieval of information was performed on PubMed, Embase, and Google Scholar. Specified keywords were used in PubMed and the articles retrieved were published in peer-reviewed scientific journals and were associated with brain GBM cancer and the sodium iodide symporter (NIS). Additionally, the words 'radionuclide therapy OR mesenchyma, OR radioiodine OR iodine-131 OR molecular imaging OR gene therapy OR translational imaging OR targeted OR theranostic OR symporter OR virus OR solid tumor OR combined therapy OR pituitary OR plasmid AND glioblastoma OR GBM OR GB OR glioma' were also used in the appropriate literature databases of PubMed and Google Scholar. A total of 68,244 articles were found in this search on Mesenchymal Stem Cell Sodium Iodide Symporter and GBM. These articles were found till 2024. To study recent advances, a filter was added to include articles only from 2014 to 2024, duplicates were removed, and articles not related to the title were excluded. These came out to be 78 articles. From these, nine were not retrieved and only seven were selected after the removal of keyword mismatched articles. Appropriate studies were isolated, and important information from each of them was understood and entered into a database from which the information was used in this article. RESULTS As a result of their natural capacity to identify malignancies, MSCs are employed as tumor therapy vehicles. Because MSCs may be transplanted using several methods, they have been proposed as the ideal vehicles for NIS gene transfer. MSCs have been used as a delivery vector for anticancer drugs in many tumor models due to their capacity to move precisely to malignancies. Also, by directly injecting radiolabeled MSCs into malignant tumors, a therapeutic dosage of beta radiation may be deposited, with the added benefit that the tumor would only localize and not spread to the surrounding healthy tissues. CONCLUSION The non-invasive imaging-based detection of glioma stem cells presents an alternate means to monitor the tumor and diagnose and evaluate recurrence. The sodium iodide symporter gene is a specific gene in a variety of human thyroid diseases that functions to move iodine into the cell. In recent years, an increasing number of studies related to the sodium iodide symporter gene have been reported in a variety of tumors and as therapeutic vectors for imaging and therapy. Gene therapy and nuclear medicine therapy for GBM provide a new direction. In all the preclinical studies reviewed, image-guided cell therapy led to greater survival benefits and, therefore, has the potential to be translated into techniques in glioblastoma treatment trials.
Collapse
Affiliation(s)
- Siddharth Shah
- Department of Neurosurgery, University of Florida, Gainesville, FL 32608, USA;
| | | |
Collapse
|
36
|
Ahmad N, Anker A, Klein S, Dean J, Knoedler L, Remy K, Pagani A, Kempa S, Terhaag A, Prantl L. Autologous Fat Grafting-A Panacea for Scar Tissue Therapy? Cells 2024; 13:1384. [PMID: 39195271 DOI: 10.3390/cells13161384] [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: 06/09/2024] [Revised: 08/04/2024] [Accepted: 08/05/2024] [Indexed: 08/29/2024] Open
Abstract
Scars may represent more than a cosmetic concern for patients; they may impose functional limitations and are frequently associated with the sensation of itching or pain, thus impacting both psychological and physical well-being. From an aesthetic perspective, scars display variances in color, thickness, texture, contour, and their homogeneity, while the functional aspect encompasses considerations of functionality, pliability, and sensory perception. Scars located in critical anatomic areas have the potential to induce profound impairments, including contracture-related mobility restrictions, thereby significantly impacting daily functioning and the quality of life. Conventional approaches to scar management may suffice to a certain extent, yet there are cases where tailored interventions are warranted. Autologous fat grafting emerges as a promising therapeutic avenue in such instances. Fundamental mechanisms underlying scar formation include chronic inflammation, fibrogenesis and dysregulated wound healing, among other contributing factors. These mechanisms can potentially be alleviated through the application of adipose-derived stem cells, which represent the principal cellular component utilized in the process of lipofilling. Adipose-derived stem cells possess the capacity to secrete proangiogenic factors such as fibroblast growth factor, vascular endothelial growth factor and hepatocyte growth factor, as well as neurotrophic factors, such as brain-derived neurotrophic factors. Moreover, they exhibit multipotency, remodel the extracellular matrix, act in a paracrine manner, and exert immunomodulatory effects through cytokine secretion. These molecular processes contribute to neoangiogenesis, the alleviation of chronic inflammation, and the promotion of a conducive milieu for wound healing. Beyond the obvious benefit in restoring volume, the adipose-derived stem cells and their regenerative capacities facilitate a reduction in pain, pruritus, and fibrosis. This review elucidates the regenerative potential of autologous fat grafting and its beneficial and promising effects on both functional and aesthetic outcomes when applied to scar tissue.
Collapse
Affiliation(s)
- Nura Ahmad
- Department of Plastic, Hand and Reconstructive Surgery, University Hospital Regensburg, Franz-Josef-Strauß Allee 11, 93053 Regensburg, Germany
| | - Alexandra Anker
- Department of Plastic, Hand and Reconstructive Surgery, University Hospital Regensburg, Franz-Josef-Strauß Allee 11, 93053 Regensburg, Germany
| | - Silvan Klein
- Department of Plastic, Hand and Reconstructive Surgery, University Hospital Regensburg, Franz-Josef-Strauß Allee 11, 93053 Regensburg, Germany
| | - Jillian Dean
- School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Leonard Knoedler
- Department of Plastic, Hand and Reconstructive Surgery, University Hospital Regensburg, Franz-Josef-Strauß Allee 11, 93053 Regensburg, Germany
| | - Katya Remy
- Division of Plastic and Reconstructive Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Andrea Pagani
- Department of Plastic, Hand and Reconstructive Surgery, University Hospital Regensburg, Franz-Josef-Strauß Allee 11, 93053 Regensburg, Germany
| | - Sally Kempa
- Department of Plastic, Hand and Reconstructive Surgery, University Hospital Regensburg, Franz-Josef-Strauß Allee 11, 93053 Regensburg, Germany
| | - Amraj Terhaag
- Department of Plastic, Hand and Reconstructive Surgery, University Hospital Regensburg, Franz-Josef-Strauß Allee 11, 93053 Regensburg, Germany
| | - Lukas Prantl
- Department of Plastic, Hand and Reconstructive Surgery, University Hospital Regensburg, Franz-Josef-Strauß Allee 11, 93053 Regensburg, Germany
| |
Collapse
|
37
|
Hajihosseintehrani M, Amini A, Heidari M, Gholipourmalekabadi M, Fadaei Fathabady F, Mostafavinia A, Ahmadi H, Khodadadi M, Naser R, Zare F, Alizadeh S, Moeinian N, Chien S, Bayat M. The Application of Photobiomodulation and Stem Cells Seeded on the Scaffold Accelerates the Wound Healing Process in Mice. J Lasers Med Sci 2024; 15:e40. [PMID: 39381785 PMCID: PMC11459249 DOI: 10.34172/jlms.2024.40] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Accepted: 05/08/2024] [Indexed: 10/10/2024]
Abstract
Introduction: The purpose of this research was to test the impact of seeding a hydrogel chitosan scaffold (HCS) with human adipose-derived stem cells (hADSCs) under the influence of photobiomodulation (PBM) on the remodeling step on the wound repairing process in mice. Methods: Thirty mice were randomly assigned to five groups (n=6 per group ): The control group (group 1) consisted of mice without any intervention. In group 2, an HCS was implanted into the wound. In group 3, a combination of HCS+hADSC was inserted into the wound. In group 4, an HCS was inserted into the wound and PBM was applied. In group 5, a combination of HCS+hADSCs was inserted into the wound, followed by PBM treatment. Results: Improvements in the injury closing rate (WCR) and microbial flora were observed in all groups. However, the highest WCRs were observed in group s 5, 4, 3, and 2 (all P values were 0.000). Groups 3-5 showed increased wound strength compared to group s 1 and 2, with group 2 demonstrating better results than group 1 (P values ranged from 0.000 to 0.013). Although group s 3-5 showed increases in certain stereological elements compared to group s 1 and 2, group 2 exhibited superior results in comparison with group 1 (P values ranged from 0.000 to 0.049). Conclusion: The joined use of HCS+hADSCs+PBM significantly accelerated the wound healing process during the maturation phase in healthy mice. This approach demonstrated superior wound healing compared to the use of HCS alone, hADSCs+HCS, or PBM+HCS. The findings suggest an additive effect when HCS+hADSCs+PBM are combined.
Collapse
Affiliation(s)
- Masoumeh Hajihosseintehrani
- Department of Biology and Anatomical Sciences at Shahid Beheshti University of Medical Sciences (SBMU), Tehran, Iran
| | - Abdollah Amini
- Department of Biology and Anatomical Sciences at Shahid Beheshti University of Medical Sciences (SBMU), Tehran, Iran
| | - Mohammadhossein Heidari
- Department of Biology and Anatomical Sciences at Shahid Beheshti University of Medical Sciences (SBMU), Tehran, Iran
| | - Mazaher Gholipourmalekabadi
- Department of Tissue Engineering & Regenerative Medicine, Iran University of Medical Sciences, Hemmat Highway, Tehran, Iran
| | - Fatemeh Fadaei Fathabady
- Department of Biology and Anatomical Sciences at Shahid Beheshti University of Medical Sciences (SBMU), Tehran, Iran
| | - Atarodalsadat Mostafavinia
- Department of Anatomical Sciences and Cognitive Neuroscience at the Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Houssein Ahmadi
- Department of Biology and Anatomical Sciences at Shahid Beheshti University of Medical Sciences (SBMU), Tehran, Iran
| | - Maryam Khodadadi
- Xi’an jiaotong University School of Stomatology, Xi’an, Shaanxi Province, China
| | - Reza Naser
- Tissue Engineering Department, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Fateme Zare
- Department of Biology and Anatomical Sciences at Shahid Beheshti University of Medical Sciences (SBMU), Tehran, Iran
| | - Sanaz Alizadeh
- Department of Anatomical Sciences and Cognitive Neuroscience at the Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Nafiseh Moeinian
- Department of Biology and Anatomical Sciences at Shahid Beheshti University of Medical Sciences (SBMU), Tehran, Iran
| | - Sufan Chien
- Price Institute of Surgical Research at the University of Louisville and Noveratech LLC of Louisville in Louisville, KY, USA
| | - Mohammad Bayat
- Department of Tissue Engineering & Regenerative Medicine, Iran University of Medical Sciences, Hemmat Highway, Tehran, Iran
- Price Institute of Surgical Research at the University of Louisville and Noveratech LLC of Louisville in Louisville, KY, USA
| |
Collapse
|
38
|
Xing T, Wang X, Xu Y, Sun F, Chen M, Yan Q, Ma Z, Jiang H, Chen X, Li X, Sultan R, Yan T, Wang Z, Jia J. Click method preserves but EDC method compromises the therapeutic activities of the peptide-activated hydrogels for critical ischemic vessel regeneration. Biomed Pharmacother 2024; 177:116959. [PMID: 38906023 DOI: 10.1016/j.biopha.2024.116959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 05/25/2024] [Accepted: 06/15/2024] [Indexed: 06/23/2024] Open
Abstract
Peptide-functionalized hydrogel is one of commonly used biomaterials to introduce hydrogel-induced vessel regeneration. Despite many reports about the discoveries of high-active peptides (or ligands) for regeneration, the study on the conjugating methods for the hydrogel functionalization with peptides is limited. Here, we compared the vasculogenic efficacy of the peptide-functionalized hydrogels prepared by two commonly used conjugating methods, 1-ethyl-3-(3-dimethylamino propyl) carbodiimide (EDC) and Click methods, through cell models, organ-on-chips models, animal models, and RNA sequencing analysis. Two vascular-related cell types, the human umbilical vein endothelial cells (HUVECs) and the adipose-derived stem cells (ADSCs), have been cultured on the hydrogel surfaces prepared by EDC/Click methods. It showed that the hydrogels prepared by Click method supported the higher vasculogenic activities while the ones made by EDC method compromised the peptide activities on hydrogels. The vasculogenesis assays further revealed that hydrogels prepared by Click method promoted a better vascular network formation. In a critical ischemic hindlimb model, only the peptide-functionalized hydrogels prepared by Click method successfully salvaged the ischemic limb, significantly improved blood perfusion, and enhanced the functional recoveries (through gait analysis and animal behavior studies). RNA sequencing studies revealed that the hydrogels prepared by Click method significantly promoted the PI3K-AKT pathway activation compared to the hydrogels prepared by EDC method. All the results suggested that EDC method compromised the functions of the peptides, while Click method preserved the vascular regenerating capacities of the peptides on the hydrogels, illustrating the importance of the conjugating method during the preparation of the peptide-functionalized hydrogels.
Collapse
Affiliation(s)
- Tongying Xing
- School of Life Sciences, Shanghai University, Shanghai, China; Sino-Swiss Institute of Advanced Technology, School of Micro-electronics, Shanghai University, Shanghai, China
| | - Xuelin Wang
- School of Life Sciences, Shanghai University, Shanghai, China; Shanghai-MOST Key Laboratory of Health and Disease Genomics, Shanghai Institute for Biomedical and Pharmaceutical Technologies (SIBPT), Shanghai, China
| | - Yongqiang Xu
- Department of colorectal surgery, The First People's Hospital of Huzhou, The First Affiliated Hospital of Huzhou University, Huzhou, Zhejiang, China
| | - Fei Sun
- School of Life Sciences, Shanghai University, Shanghai, China
| | - Min Chen
- School of Life Sciences, Shanghai University, Shanghai, China
| | - Qiang Yan
- Department of Surgery, Huzhou Central Hospital, Huzhou, Zhejiang, China; Department of Surgery, Affiliated Central Hospital Huzhou University, Huzhou, Zhejiang, China
| | - Zhihong Ma
- Department of Precision Medical Clinical Research Center, Huzhou Central Hospital, Huzhou, Zhejiang, China
| | - Haihong Jiang
- School of Life Sciences, Shanghai University, Shanghai, China
| | - Xingxing Chen
- School of Life Sciences, Shanghai University, Shanghai, China
| | - Xueyi Li
- Sino-Swiss Institute of Advanced Technology, School of Micro-electronics, Shanghai University, Shanghai, China
| | - Rabia Sultan
- School of Life Sciences, Shanghai University, Shanghai, China
| | - Tingdong Yan
- School of Life Sciences, Shanghai University, Shanghai, China.
| | - Zhimin Wang
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, Shanghai Institute for Biomedical and Pharmaceutical Technologies (SIBPT), Shanghai, China.
| | - Jia Jia
- School of Life Sciences, Shanghai University, Shanghai, China; Sino-Swiss Institute of Advanced Technology, School of Micro-electronics, Shanghai University, Shanghai, China.
| |
Collapse
|
39
|
Foti R, Storti G, Palmesano M, Scioli MG, Fiorelli E, Terriaca S, Cervelli G, Kim BS, Orlandi A, Cervelli V. Senescence in Adipose-Derived Stem Cells: Biological Mechanisms and Therapeutic Challenges. Int J Mol Sci 2024; 25:8390. [PMID: 39125960 PMCID: PMC11312747 DOI: 10.3390/ijms25158390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Revised: 07/24/2024] [Accepted: 07/25/2024] [Indexed: 08/12/2024] Open
Abstract
Adipose tissue-derived stem cells (ADSCs) represent a subset of the mesenchymal stem cells in every adipose compartment throughout the body. ADSCs can differentiate into various cell types, including chondrocytes, osteocytes, myocytes, and adipocytes. Moreover, they exhibit a notable potential to differentiate in vitro into cells from other germinal lineages, including endothelial cells and neurons. ADSCs have a wide range of clinical applications, from breast surgery to chronic wounds. Furthermore, they are a promising cell population for future tissue-engineering uses. Accumulating evidence indicates a decreased proliferation and differentiation potential of ADSCs with an increasing age, increasing body mass index, diabetes mellitus, metabolic syndrome, or exposure to radiotherapy. Therefore, the recent literature thoroughly investigates this cell population's senescence mechanisms and how they can hinder its possible therapeutic applications. This review will discuss the biological mechanisms and the physio-pathological causes behind ADSC senescence and how they can impact cellular functionality. Moreover, we will examine the possible strategies to invert these processes, re-establishing the full regenerative potential of this progenitor population.
Collapse
Affiliation(s)
- Riccardo Foti
- Plastic Surgery, Department of Surgical Sciences, University of Rome “Tor Vergata”, 00133 Rome, Italy; (R.F.); (M.P.); (V.C.)
| | - Gabriele Storti
- Plastic Surgery, Department of Surgical Sciences, University of Rome “Tor Vergata”, 00133 Rome, Italy; (R.F.); (M.P.); (V.C.)
| | - Marco Palmesano
- Plastic Surgery, Department of Surgical Sciences, University of Rome “Tor Vergata”, 00133 Rome, Italy; (R.F.); (M.P.); (V.C.)
| | - Maria Giovanna Scioli
- Anatomy Pathology Institute, Department of Biomedicine and Prevention, University of Rome “Tor Vergata”, 00133 Rome, Italy; (M.G.S.); (E.F.); (S.T.); (A.O.)
| | - Elena Fiorelli
- Anatomy Pathology Institute, Department of Biomedicine and Prevention, University of Rome “Tor Vergata”, 00133 Rome, Italy; (M.G.S.); (E.F.); (S.T.); (A.O.)
| | - Sonia Terriaca
- Anatomy Pathology Institute, Department of Biomedicine and Prevention, University of Rome “Tor Vergata”, 00133 Rome, Italy; (M.G.S.); (E.F.); (S.T.); (A.O.)
| | - Giulio Cervelli
- Department of Experimental Medicine, University of Rome “Tor Vergata”, 00133 Rome, Italy;
| | - Bong Sung Kim
- Department of Plastic Surgery and Hand Surgery, University Hospital Zurich, 8006 Zurich, Switzerland;
| | - Augusto Orlandi
- Anatomy Pathology Institute, Department of Biomedicine and Prevention, University of Rome “Tor Vergata”, 00133 Rome, Italy; (M.G.S.); (E.F.); (S.T.); (A.O.)
| | - Valerio Cervelli
- Plastic Surgery, Department of Surgical Sciences, University of Rome “Tor Vergata”, 00133 Rome, Italy; (R.F.); (M.P.); (V.C.)
| |
Collapse
|
40
|
Ferro AP, de Jesus Guirro RR, Orellana MD, de Santis GC, Farina Junior JA, de Oliveira Guirro EC. Photobiomodulation with laser and led on mesenchymal stem cells viability and wound closure in vitro. Lasers Med Sci 2024; 39:205. [PMID: 39088075 DOI: 10.1007/s10103-024-04159-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 07/25/2024] [Indexed: 08/02/2024]
Abstract
Mesenchymal stem cells can differentiate into specific cell lineages in the tissue repair process. Photobiomodulation with laser and LED is used to treat several comorbidities, can interfere in cell proliferation and viability, in addition to promoting responses related to the physical parameters adopted. Evaluate and compare the effects of laser and LED on mesenchymal cells, with different energy doses and different wavelengths, in addition to viability and wound closure. Mesenchymal stem cells derived from human adipocytes were irradiated with laser (energy of 0.5 J, 2 J and 4 J, wavelength of 660 nm and 830 nm), and LED (energy of 0.5 J, 2 J and 4 J, where lengths are 630 nm and 850 nm). The wound closure process was evaluated through monitoring the reduction of the lesion area in vitro. Viability was determined by analysis with Hoechst and Propidium Iodide markers, and quantification of viable and non-viable cells respectively Data distributions were analyzed using the Shapiro-Wilk test. Homogeneity was analyzed using Levene's test. The comparison between the parameters used was analyzed using the Two-way ANOVA test. The T test was applied to data relating to viability and lesion area. For LED photobiomodulation, only the 630 nm wavelength obtained a significant result in 24, 48 and 72 h (p = 0,027; p = 0,024; p = 0,009). The results related to the in vitro wound closure test indicate that both photobiomodulation with laser and LED demonstrated significant results considering the time it takes to approach the edges (p < 0.05). Considering the in vitro experimental conditions of the study, it is possible to conclude that the physical parameters of photobiomodulation, such as energy and wavelength, with laser or LED in mesenchymal stem cells, can play a potential role in cell viability and wound closure.
Collapse
Affiliation(s)
- Ana Paula Ferro
- Department of Health Sciences, Postgraduate Program in Rehabilitation and Functional Performance, Ribeirão Preto Medical School, University of São Paulo, Bandeirantes Avenue, 3900, Monte Alegre, CEP 14049-900, Ribeirão Preto, São Paulo, Brazil.
| | - Rinaldo Roberto de Jesus Guirro
- Department of Health Sciences, Postgraduate Program in Rehabilitation and Functional Performance, Ribeirão Preto Medical School, University of São Paulo, Bandeirantes Avenue, 3900, Monte Alegre, CEP 14049-900, Ribeirão Preto, São Paulo, Brazil
| | - Maristela Delgado Orellana
- Cell Biology Laboratory, Ribeirão Preto Blood Center Foundation, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Gil Cunha de Santis
- Cell Therapy Laboratory, Regional Blood Center of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Jayme Adriano Farina Junior
- Department of Surgery, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Elaine Caldeira de Oliveira Guirro
- Department of Health Sciences, Postgraduate Program in Rehabilitation and Functional Performance, Ribeirão Preto Medical School, University of São Paulo, Bandeirantes Avenue, 3900, Monte Alegre, CEP 14049-900, Ribeirão Preto, São Paulo, Brazil
| |
Collapse
|
41
|
Yari F, Ashoub MH, Amirizadeh N, Nikougoftar M, Valandani HM, Khalilabadi RM. Differential Expression of the hTERT Gene in Umbilical Cord-Derived Mesenchymal Stem Cells Cocultured with B Cell Precursor Leukemia Cell Microparticles or CD41 +/CD61 + Platelet Microparticles. Biochem Genet 2024; 62:2796-2809. [PMID: 38019337 DOI: 10.1007/s10528-023-10565-9] [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: 05/17/2023] [Accepted: 10/26/2023] [Indexed: 11/30/2023]
Abstract
Several investigations are being done to increase the short lifetime of mesenchymal stem cells (MSCs). One of the crucial genes involved in the immortalization of MSCs, hTERT (human telomerase reverse transcriptase), is activated in most publications using viral-based techniques. In this work, we investigated the use of platelet-derived (PMPs) and B cell precursor leukemia-derived microparticles as a nonviral method to trigger and compare the expression of the hTERT gene in MSCs. MSCs were extracted from the umbilical cord for the current investigation and identified using a flow cytometry approach and an inverted microscope. The Nalm-6 cell line and platelet concentrate were used to isolate microparticles (MPs). MSCs and MPs were cocultured for 14 days at 25-, 50-, and 100 μg/ml concentrations. qRT-PCR was used to research the expression of the hTERT gene. SPSS 26.0's t test was used to compare the outcomes. After coculture with platelet MPs, MSCs had higher levels of hTERT gene expression than the control group. In contrast, this gene's expression was concurrently decreased in MSCs exposed to MPs generated from Nalm-6. We demonstrated that following 14-day treatment, PMP significantly boosted the hTERT gene expression in MSCs, while the Nalm-6 MPs lowered the gene expression. However, additional studies are necessary due to the stability of hTERT gene expression and the immortalization of MSCs following exposure.
Collapse
Affiliation(s)
- Fatemeh Yari
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| | - Muhammad Hossein Ashoub
- Department of Hematology and Medical Laboratory Sciences, Faculty of Allied Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Naser Amirizadeh
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| | - Mahin Nikougoftar
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| | - Hajar Mardani Valandani
- Department of Hematology and Medical Laboratory Sciences, Faculty of Allied Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Roohollah Mirzaee Khalilabadi
- Department of Hematology and Medical Laboratory Sciences, Faculty of Allied Medicine, Kerman University of Medical Sciences, Kerman, Iran.
| |
Collapse
|
42
|
Ahmadieh-Yazdi A, Karimi M, Afkhami E, Hajizadeh-Tafti F, Kuchakzadeh F, Yang P, Sheykhhasan M. Unveiling therapeutic potential: Adipose tissue-derived mesenchymal stem cells and their exosomes in the management of diabetes mellitus, wound healing, and chronic ulcers. Biochem Pharmacol 2024; 226:116399. [PMID: 38944396 DOI: 10.1016/j.bcp.2024.116399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 05/30/2024] [Accepted: 06/26/2024] [Indexed: 07/01/2024]
Abstract
Diabetes mellitus (DM) is a pervasive global health issue with substantial morbidity and mortality, often resulting in secondary complications, including diabetic wounds (DWs). These wounds, arising from hyperglycemia, diabetic neuropathy, anemia, and ischemia, afflict approximately 15% of diabetic patients, with a considerable 25% at risk of lower limb amputations. The conventional approaches for chronic and diabetic wounds management involves utilizing various therapeutic substances and techniques, encompassing growth factors, skin substitutes and wound dressings. In parallel, emerging cell therapy approaches, notably involving adipose tissue-derived mesenchymal stem cells (ADMSCs), have demonstrated significant promise in addressing diabetes mellitus and its complications. ADMSCs play a pivotal role in wound repair, and their derived exosomes have garnered attention for their therapeutic potential. This review aimed to unravel the potential mechanisms and provide an updated overview of the role of ADMSCs and their exosomes in diabetes mellitus and its associated complications, with a specific focus on wound healing.
Collapse
Affiliation(s)
- Amirhossein Ahmadieh-Yazdi
- Stem Cell Biology Research Center, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Mahdieh Karimi
- Stem Cell Biology Research Center, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Elham Afkhami
- Stem Cell Biology Research Center, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran; Department of Anatomical Sciences, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Fatemeh Hajizadeh-Tafti
- Stem Cell Biology Research Center, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Fatemeh Kuchakzadeh
- Stem Cell Biology Research Center, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran; Department of Tissue Engineering, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Piao Yang
- Department of Molecular Genetics, College of Arts and Sciences, The Ohio State University, Columbus, OH 43210, USA
| | - Mohsen Sheykhhasan
- Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran.
| |
Collapse
|
43
|
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
|
44
|
He Y, Tang Y, Zeng B, Chen X, Yuan L, Lu Y, Du W, Li R, Han Y, Deng F, Yu D, Zhao W. Black phosphorus quantum dot-modified ADSCs as a novel therapeutic for periodontitis bone loss coupling of osteogenesis and osteoimmunomodulation. Mater Today Bio 2024; 27:101122. [PMID: 38975241 PMCID: PMC11225909 DOI: 10.1016/j.mtbio.2024.101122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 05/24/2024] [Accepted: 06/09/2024] [Indexed: 07/09/2024] Open
Abstract
Alveolar bone defect repair remains a persistent clinical challenge for periodontitis treatment. The use of peripheral functional seed cells is a hot topic in periodontitis. Herein, we explored the cellular behaviors and osteogenic ability of adipose-derived mesenchymal stem cells (ADSCs) treated with black phosphorus quantum dots (BPQDs). Additionally, macrophage polarization, osteogenic effects and angiogenesis were investigated through the paracrine pathway regulated by BPQD-modified ADSCs. Our results demonstrated that BPQDs showed good biocompatibility with ADSCs and BPQD-modified ADSCs could improve the bone repair in vivo inflammatory microenvironment by regulating osteogenesis and osteoimmunomodulation. The BPQDs increased the osteogenic differentiation of ADSCs via the Wnt/β-catenin and BMP2/SMAD5/Runx2 signaling pathway. In addition, BPQD-modified ADSCs promoted the osteogenic effect of BMSCs and facilitated the polarization of macrophages from M1 towards M2 phenotype transformation through the paracrine pathway in the periodontitis microenvironment. This strategy provides a novel idea for treatment of alveolar bone defects for periodontitis in the foreseeable future.
Collapse
Affiliation(s)
- Yi He
- Hospital of Stomatology, Guanghua School of Stomatology, Institute of Stomatological Research, Sun Yat-sen University, Guangzhou 510080, China
- Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou 510080, China
| | - Yuquan Tang
- Zhujiang Hospital, Southern Medical University, Guangzhou, 510080, China
| | - Binghui Zeng
- Hospital of Stomatology, Guanghua School of Stomatology, Institute of Stomatological Research, Sun Yat-sen University, Guangzhou 510080, China
- Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou 510080, China
| | - Xun Chen
- Hospital of Stomatology, Guanghua School of Stomatology, Institute of Stomatological Research, Sun Yat-sen University, Guangzhou 510080, China
- Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou 510080, China
| | - Linyu Yuan
- Hospital of Stomatology, Guanghua School of Stomatology, Institute of Stomatological Research, Sun Yat-sen University, Guangzhou 510080, China
- Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou 510080, China
| | - Yunyang Lu
- Hospital of Stomatology, Guanghua School of Stomatology, Institute of Stomatological Research, Sun Yat-sen University, Guangzhou 510080, China
- Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou 510080, China
| | - Weidong Du
- Hospital of Stomatology, Guanghua School of Stomatology, Institute of Stomatological Research, Sun Yat-sen University, Guangzhou 510080, China
- Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou 510080, China
| | - Runze Li
- Hospital of Stomatology, Guanghua School of Stomatology, Institute of Stomatological Research, Sun Yat-sen University, Guangzhou 510080, China
- Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou 510080, China
| | - Yaolin Han
- Hospital of Stomatology, Guanghua School of Stomatology, Institute of Stomatological Research, Sun Yat-sen University, Guangzhou 510080, China
- Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou 510080, China
| | - Feilong Deng
- Hospital of Stomatology, Guanghua School of Stomatology, Institute of Stomatological Research, Sun Yat-sen University, Guangzhou 510080, China
- Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou 510080, China
| | - Dongsheng Yu
- Hospital of Stomatology, Guanghua School of Stomatology, Institute of Stomatological Research, Sun Yat-sen University, Guangzhou 510080, China
- Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou 510080, China
| | - Wei Zhao
- Hospital of Stomatology, Guanghua School of Stomatology, Institute of Stomatological Research, Sun Yat-sen University, Guangzhou 510080, China
- Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou 510080, China
| |
Collapse
|
45
|
Prakashan D, Singh A, Deshpande AD, Chandra V, Sharma GT, Gandhi S. Bone marrow derived mesenchymal stem cells enriched PCL-gelatin nanofiber scaffold for improved wound healing. Int J Biol Macromol 2024; 274:133447. [PMID: 38944073 DOI: 10.1016/j.ijbiomac.2024.133447] [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: 03/26/2024] [Revised: 06/04/2024] [Accepted: 06/24/2024] [Indexed: 07/01/2024]
Abstract
Electrospun nanofibers exhibit a significant potential in the synthesis of nanostructured materials, thereby offering a promising avenue for enhancing the efficacy of wound care. The present study aimed to investigate the wound-healing potential of two biomacromolecules, PCL-Gelatin nanofiber adhered with bone marrow-derived mesenchymal stem cells (BMSCs). Characterisation of the nanofiber revealed a mean fiber diameter ranging from 200 to 300 nm, with distinctive elemental peaks corresponding to polycaprolactone (PCL) and gelatin. Additionally, BMSCs derived from bone marrow were integrated into nanofibers, and their wound-regenerative potential was systematically evaluated through both in-vitro and in-vivo methodologies. In-vitro assessments substantiated that BMSC-incorporated nanofibers enhanced cell viability and crucial cellular processes such as adhesion, and proliferation. Subsequently, in-vivo studies were performed to demonstrate the wound-healing efficacy of nanofibers. It was observed that the rate of wound healing of BMSCs incorporated nanofibers surpassed both, nanofiber and BMSCs alone. Furthermore, histomorphological analysis revealed accelerated re-epithelization and improved wound contraction in BMSCs incorporated nanofiber group. The fabricated nanofiber incorporated with BMSCs exhibited superior wound regeneration in animal model and may be utilised as a wound healing patch.
Collapse
Affiliation(s)
- Drishya Prakashan
- DBT-National Institute of Animal Biotechnology (NIAB), Hyderabad-500032, Telangana, India; DBT-Regional Centre for Biotechnology (RCB), Faridabad-121001, Haryana, India
| | - Archita Singh
- ICAR-Indian Veterinary Research Institute, Izatnagar-243122, U.P., India
| | - Aditya D Deshpande
- DBT-National Institute of Animal Biotechnology (NIAB), Hyderabad-500032, Telangana, India; ICAR-Indian Veterinary Research Institute, Izatnagar-243122, U.P., India
| | - Vikash Chandra
- ICAR-Indian Veterinary Research Institute, Izatnagar-243122, U.P., India
| | - G Taru Sharma
- DBT-National Institute of Animal Biotechnology (NIAB), Hyderabad-500032, Telangana, India; DBT-Regional Centre for Biotechnology (RCB), Faridabad-121001, Haryana, India.
| | - Sonu Gandhi
- DBT-National Institute of Animal Biotechnology (NIAB), Hyderabad-500032, Telangana, India; DBT-Regional Centre for Biotechnology (RCB), Faridabad-121001, Haryana, India.
| |
Collapse
|
46
|
Rodrigues GM, de Almeida ME, Marcelino SAC, Fernandes PBU, da Cruz JOP, Araújo FL, Ferreira RDS, Botelho AFM, Bedoya FJ, Cahuana GM, Hitos AB, Soria B, Costal-Oliveira F, Duarte CG, Tejedo JR, Chávez-Olórtegui C, Melo MM. Protective effects of mesenchymal stromal cell-derived secretome on dermonecrosis induced in rabbits by Loxosceles intermedia spider venom. J Venom Anim Toxins Incl Trop Dis 2024; 30:e20240004. [PMID: 39069986 PMCID: PMC11276892 DOI: 10.1590/1678-9199-jvatitd-2024-0004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 06/21/2024] [Indexed: 07/30/2024] Open
Abstract
Background Loxoscelism refers to a set of clinical manifestations caused by the bite of spiders from the Loxosceles genus. The classic clinical symptoms are characterized by an intense inflammatory reaction at the bite site followed by local necrosis and can be classified as cutaneous loxoscelism. This cutaneous form presents difficult healing, and the proposed treatments are not specific or effective. This study aimed to evaluate the protective effect of mesenchymal stromal cells-derived secretome on dermonecrosis induced by Loxosceles intermedia spider venom in rabbits. Methods Sixteen rabbits were distributed into four groups (n = 4). Except for group 1 (G1), which received only PBS, the other three groups (G2, G3, and G4) were initially challenged with 10 μg of L. intermedia venom, diluted in 100 μL of NaCl 0.9%, by intradermic injection in the interscapular region. Thirty minutes after the challenge all groups were treated with secretome, except for group 2. Group 1 (G1-control group) received intradermal injection (ID) of 60 μg of secretome in 0.15 M PBS; Group 2 (G2) received 0.9% NaCl via ID; Group 3 (G3) received 60 μg of secretome, via ID and Group 4 (G4), received 60 μg of secretome by intravenous route. Rabbits were evaluated daily and after 15 days were euthanized, necropsied and skin samples around the necrotic lesions were collected for histological analysis. Results Rabbits of G1 did not present edema, erythema, hemorrhagic halo, or necrosis. In animals from G2, G3, and G4, edema appeared after 6h. However, minor edema was observed in the animals of G2 and G3. Hemorrhagic halo was observed in animals, six hours and three days after, on G2, G3, and G4. Macroscopically, in G4, only one animal out of four had a lesion that evolved into a dermonecrotic wound. No changes were observed in the skin of the animals of G1, by microscopic evaluation. All animals challenged with L. intermedia venom showed similar alterations, such as necrosis and heterophilic infiltration. However, animals from G4 showed fibroblast activation, early development of connective tissue, neovascularization, and tissue re-epithelialization, indicating a more prominent healing process. Conclusion These results suggest that secretome from mesenchymal stromal cells cultured in a xeno-free and human component-free culture media can be promising to treat dermonecrosis caused after Loxosceles spiders bite envenoming.
Collapse
Affiliation(s)
- Gabriela Marques Rodrigues
- Department of Veterinary Clinic and Surgery, Veterinary College,
Federal University of Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Mara Elvira de Almeida
- Department of Veterinary Clinic and Surgery, Veterinary College,
Federal University of Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Sóstenes Apolo Correia Marcelino
- Department of Veterinary Clinic and Surgery, Veterinary College,
Federal University of Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Paula Bretas Ullmann Fernandes
- Department of Veterinary Clinic and Surgery, Veterinary College,
Federal University of Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Jessica Oliveira Pereira da Cruz
- Department of Veterinary Clinic and Surgery, Veterinary College,
Federal University of Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Françoise Louanne Araújo
- Department of Veterinary Clinic and Surgery, Veterinary College,
Federal University of Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Raquel da Silva Ferreira
- Department of Veterinary Clinic and Surgery, Veterinary College,
Federal University of Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Ana Flávia Machado Botelho
- Department of Veterinary Clinic and Surgery, Veterinary College,
Federal University of Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Francisco Javier Bedoya
- Department of Molecular Biology and Biochemical Engineering,
Universidad Pablo de Olavide, Seville, Spain
- Biomedical Research Network for Diabetes and Related Metabolic
Diseases (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| | - Gladys Margot Cahuana
- Department of Molecular Biology and Biochemical Engineering,
Universidad Pablo de Olavide, Seville, Spain
- Biomedical Research Network for Diabetes and Related Metabolic
Diseases (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| | - Ana Belén Hitos
- Institute of Bioengineering and Institute of Biomedical Research
ISABIAL, University Miguel Hernández de Elche, Alicante, Spain
| | - Bernat Soria
- Biomedical Research Network for Diabetes and Related Metabolic
Diseases (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
- Institute of Bioengineering and Institute of Biomedical Research
ISABIAL, University Miguel Hernández de Elche, Alicante, Spain
| | - Fernanda Costal-Oliveira
- Department of Biochemistry and Immunology, Institute of Biological
Sciences, Federal University of Minas Gerais (UFMG), Belo Horizonte, MG,
Brazil
| | | | - Juan R. Tejedo
- Department of Molecular Biology and Biochemical Engineering,
Universidad Pablo de Olavide, Seville, Spain
- Biomedical Research Network for Diabetes and Related Metabolic
Diseases (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
- Institute of Tropical Diseases, Universidad Nacional Toribio
Rodríguez de Mendoza de Amazonas, Chachapoyas, Peru
| | - Carlos Chávez-Olórtegui
- Department of Biochemistry and Immunology, Institute of Biological
Sciences, Federal University of Minas Gerais (UFMG), Belo Horizonte, MG,
Brazil
| | - Marília Martins Melo
- Department of Veterinary Clinic and Surgery, Veterinary College,
Federal University of Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| |
Collapse
|
47
|
Liang Q, Zhou D, Ge X, Song P, Chu W, Xu J, Shen Y. Exosomes from adipose-derived mesenchymal stem cell improve diabetic wound healing and inhibit fibrosis via miR-128-1-5p/TGF-β1/Smad axis. Mol Cell Endocrinol 2024; 588:112213. [PMID: 38556162 DOI: 10.1016/j.mce.2024.112213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Revised: 03/13/2024] [Accepted: 03/17/2024] [Indexed: 04/02/2024]
Abstract
OBJECTIVE Difficult-to-heal wound is a prevalent and significant complication of diabetes, characterized by impaired functionality of epithelial cells such as fibroblasts. This study aims to investigate the potential mechanism of ADSC-Exos promoting diabetic wound healing by regulating fibroblast function. MATERIALS AND METHODS ADSC-Exos were confirmed through TEM, NTA, and Western Blot techniques. The study conducted on rat skin fibroblasts (RSFs) exposed to 33 mmol/L glucose in vitro. We used cck-8, EDU, transwell, and scratch assays to verify the proliferation and migration of RSFs. Furthermore, levels of TGF-β1 and α-SMA proteins were determined by immunofluorescence and Western Blot. RSFs were transfected with miR-128-1-5p mimics and inhibitors, followed by quantification of TGF-β1, α-SMA, Col I and Smad2/3 protein levels using Western Blot. In vivo, the effects of ADSC-Exos on diabetic wounds were assessed using digital imaging, histological staining, as well as Western Blot analysis. RESULTS In vitro, ADSC-Exos significantly enhanced proliferation and migration of RSFs while reducing the expression of TGF-β1 and α-SMA. In vivo, ADSC-Exos effectively promoted diabetic wound healing and mitigated scar fibrosis. Additionally, ADSC-Exos exhibited elevated levels of miR-128-1-5p, which targets TGF-β1, resulting in a notable reduction in TGF-β1, α-SMA, Col I and smad2/3 phosphorylation in RSFs. CONCLUSION In conclusion, our results demonstrated that ADSC-Exos promoted diabetic wound healing, and inhibited skin fibrosis by regulating miR-128-1-5p/TGF-β1/Smad signaling pathway, which provides a promising innovative treatment for diabetic wound healing.
Collapse
Affiliation(s)
- Qiu Liang
- Department of Plastic Surgery and Burn, The First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, 233000, China; Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical University, Bengbu, Anhui, 233000, China
| | - Danlian Zhou
- Department of Plastic Surgery and Burn, The First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, 233000, China; Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical University, Bengbu, Anhui, 233000, China
| | - Xiuyu Ge
- Department of Plastic Surgery and Burn, The First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, 233000, China; Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical University, Bengbu, Anhui, 233000, China
| | - Peijun Song
- Department of Plastic Surgery and Burn, The First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, 233000, China
| | - Weiwei Chu
- Department of Plastic Surgery and Burn, The First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, 233000, China
| | - Jing Xu
- Department of Plastic Surgery and Burn, The First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, 233000, China; Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical University, Bengbu, Anhui, 233000, China.
| | - Yan Shen
- Department of Prevention and Health Care, The First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, 233000, China.
| |
Collapse
|
48
|
Liu ZX, Liu GQ, Lin ZX, Chen YQ, Chen P, Hu YJ, Yu B, Jiang N. Effects of Staphylococcus aureus on stem cells and potential targeted treatment of inflammatory disorders. Stem Cell Res Ther 2024; 15:187. [PMID: 38937829 PMCID: PMC11210046 DOI: 10.1186/s13287-024-03781-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 06/02/2024] [Indexed: 06/29/2024] Open
Abstract
Due to the advanced studies on stem cells in developmental biology, the roles of stem cells in the body and their phenotypes in related diseases have not been covered clearly. Meanwhile, with the intensive research on the mechanisms of stem cells in regulating various diseases, stem cell therapy is increasingly being attention because of its effectiveness and safety. As one of the most widely used stem cell in stem cell therapies, hematopoietic stem cell transplantation shows huge advantage in treatment of leukemia and other blood-malignant diseases. Besides, due to the effect of anti-inflammatory and immunomodulatory, mesenchymal stem cells could be a potential therapeutic strategy for variety infectious diseases. In this review, we summarized the effects of Staphylococcus aureus (S. aureus) and its components on different types of adult stem cells and their downstream signaling pathways. Also, we reviewed the roles of different kinds of stem cells in various disease models caused by S. aureus, providing new insights for applying stem cell therapy to treat infectious diseases.
Collapse
Affiliation(s)
- Zi-Xian Liu
- Division of Orthopaedics & Traumatology, Department of Orthopaedics, Southern Medical University Nanfang Hospital, Guangzhou, 510515, China
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Southern Medical University Nanfang Hospital, Guangzhou, 510515, China
- Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou, 730000, China
| | - Guan-Qiao Liu
- Division of Orthopaedics & Traumatology, Department of Orthopaedics, Southern Medical University Nanfang Hospital, Guangzhou, 510515, China
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Southern Medical University Nanfang Hospital, Guangzhou, 510515, China
| | - Ze-Xin Lin
- Division of Orthopaedics & Traumatology, Department of Orthopaedics, Southern Medical University Nanfang Hospital, Guangzhou, 510515, China
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Southern Medical University Nanfang Hospital, Guangzhou, 510515, China
| | - Ying-Qi Chen
- Division of Orthopaedics & Traumatology, Department of Orthopaedics, Southern Medical University Nanfang Hospital, Guangzhou, 510515, China
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Southern Medical University Nanfang Hospital, Guangzhou, 510515, China
| | - Peng Chen
- Division of Orthopaedics & Traumatology, Department of Orthopaedics, Southern Medical University Nanfang Hospital, Guangzhou, 510515, China
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Southern Medical University Nanfang Hospital, Guangzhou, 510515, China
| | - Yan-Jun Hu
- Division of Orthopaedics & Traumatology, Department of Orthopaedics, Southern Medical University Nanfang Hospital, Guangzhou, 510515, China
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Southern Medical University Nanfang Hospital, Guangzhou, 510515, China
| | - Bin Yu
- Division of Orthopaedics & Traumatology, Department of Orthopaedics, Southern Medical University Nanfang Hospital, Guangzhou, 510515, China.
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Southern Medical University Nanfang Hospital, Guangzhou, 510515, China.
| | - Nan Jiang
- Division of Orthopaedics & Traumatology, Department of Orthopaedics, Southern Medical University Nanfang Hospital, Guangzhou, 510515, China.
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Southern Medical University Nanfang Hospital, Guangzhou, 510515, China.
| |
Collapse
|
49
|
Wang K, Yang Z, Zhang B, Gong S, Wu Y. Adipose-Derived Stem Cell Exosomes Facilitate Diabetic Wound Healing: Mechanisms and Potential Applications. Int J Nanomedicine 2024; 19:6015-6033. [PMID: 38911504 PMCID: PMC11192296 DOI: 10.2147/ijn.s466034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 06/08/2024] [Indexed: 06/25/2024] Open
Abstract
Wound healing in diabetic patients is frequently hampered. Adipose-derived stem cell exosomes (ADSC-eoxs), serving as a crucial mode of intercellular communication, exhibit promising therapeutic roles in facilitating wound healing. This review aims to comprehensively outline the molecular mechanisms through which ADSC-eoxs enhance diabetic wound healing. We emphasize the biologically active molecules released by these exosomes and their involvement in signaling pathways associated with inflammation modulation, cellular proliferation, vascular neogenesis, and other pertinent processes. Additionally, the clinical application prospects of the reported ADSC-eoxs are also deliberated. A thorough understanding of these molecular mechanisms and potential applications is anticipated to furnish a theoretical groundwork for combating diabetic wound healing.
Collapse
Affiliation(s)
- Kang Wang
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Zihui Yang
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Boyu Zhang
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Song Gong
- Division of Endocrinology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Yiping Wu
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| |
Collapse
|
50
|
Rezaee Asl RS, Rahimzadeh-Bajgiran F, Saburi E. Evaluation of osteoconductive effect of polycaprolactone (PCL) scaffold treated with fibronectin on adipose-derived mesenchymal stem cells (AD-MSCs). AMERICAN JOURNAL OF STEM CELLS 2024; 13:152-161. [PMID: 39021375 PMCID: PMC11249668 DOI: 10.62347/dmky5924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 06/11/2024] [Indexed: 07/20/2024]
Abstract
BACKGROUND Replacing damaged organs or tissues and repairing damage by tissue engineering are attracting great interest today. A potentially effective method for bone remodeling involves combining nanofiber scaffolds with extracellular matrix (ECM), and growth factors. Today, electrospun PCL-based scaffolds are widely used for tissue engineering applications. METHODS In this study, we used an electrospun polycaprolactone (PCL) scaffold coated with fibronectin (Fn), a ubiquitous ECM glycoprotein, to investigate the induction potential of this scaffold in osteogenesis with adipose-derived mesenchymal stem cells (AD-MSCs). RESULTS Scanning electron microscopy (SEM) analysis showed that fibronectin, by binding to the membrane receptors of mesenchymal stem cells (MSCs), leads to their attachment and proliferation on the PCL scaffold and provides a suitable environment for osteogenesis. In addition, biochemical tests showed that fibronectin leads to increased calcium deposition. The results also showed that alkaline phosphatase activity was significantly higher in the PCL scaffold coated with fibronectin than in the control groups (PCL scaffold group and tissue culture polystyrene (TCPS) group) (P<0.05). Also, the analysis of quantitative reverse transcription PCR (qRT-PCR) data showed that the relative expression of bone marker genes such as osteonectin (ON), osteocalcin (OC), RUNX family transcription factor 2 (RUNX2), and collagen type I alpha 1 (COL1) was much higher in the cells seeded on the PCL/Fn scaffold than in the other groups (P<0.05). CONCLUSIONS The results show that fibronectin has an increasing effect in accelerating bone formation and promising potential for use in bone tissue engineering.
Collapse
Affiliation(s)
| | | | - Ehsan Saburi
- Medical Genetics Research Center, Mashhad University of Medical SciencesMashhad, Iran
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical SciencesMashhad, Iran
| |
Collapse
|