1
|
Tian Y, Zhang J, Jia Z, Pan X, Hu Z, Kang R, Zhou X, Luo L, Shen Z, Shen Q. Biomimetic mineralized mesenchymal stem cell-derived exosomes for dual modulation of ferroptosis and lactic acid-driven inflammation in acute liver injury therapy. J Colloid Interface Sci 2025; 687:489-506. [PMID: 39970589 DOI: 10.1016/j.jcis.2025.02.078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 02/12/2025] [Accepted: 02/13/2025] [Indexed: 02/21/2025]
Abstract
Acute liver injury (ALI) is characterized by rapid and severe hepatocellular damage, leading to ferroptosis and an exacerbated inflammatory response. Mesenchymal stem cell-derived exosomes (MSC-exo) have emerged as a promising therapeutic strategy for ALI due to their ability to deliver antioxidants and stabilize solute carrier family 7 members 11 (SLC7A11)/glutathione peroxidase 4 (GPX4) system. In this study, we developed a novel engineered exosome, MSC-exo/MnO2@DEX, by encapsulating the anti-inflammatory drug dexamethasone (DEX) within MSC-exo and modifying its surface with manganese dioxide (MnO2) via a bionano-mineralization approach. MnO2 exhibits multi-enzymatic activity, enabling efficient scavenging of reactive oxygen species (ROS), such as hydrogen peroxide and superoxide anions. When combined with MSC-exo, MnO2 not only reduces ROS levels and generates oxygen but also stabilizes the SLC7A11/GPX4 axis, thereby protecting hepatocytes from ferroptosis. Concurrently, DEX suppresses the nuclear factor-κB (NF-κB) signaling pathway, inhibits macrophage M1 polarization, and alleviates hepatic inflammation. The oxygen produced by MnO2 catalysis further mitigates hypoxia, decreases lactic acid accumulation, and downregulates histone lactylation, synergizing with DEX to enhance NF-κB pathway inhibition and amplify anti-inflammatory effects. Transcriptomic analyses revealed that MSC-exo/MnO2@DEX significantly enhances antioxidant capacity, metabolic processes, and immune function, while improving liver function and suppressing ferroptosis, lactylation and inflammatory responses. Collectively, these findings demonstrate the therapeutic potential of MSC-exo/MnO2@DEX as an effective treatment for ALI.
Collapse
Affiliation(s)
- Yiwei Tian
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| | - Jun Zhang
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| | - Zengguang Jia
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| | - Xiuhua Pan
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| | - Zongwei Hu
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| | - Ruixin Kang
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| | - Xiawei Zhou
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| | - Lin Luo
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| | - Ziqi Shen
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| | - Qi Shen
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China.
| |
Collapse
|
2
|
Suchanecka M, Grzelak J, Farzaneh M, Azizidoost S, Dari MAG, Józkowiak M, Data K, Domagała D, Niebora J, Kotrych K, Czerny B, Kamiński A, Torlińska-Walkowiak N, Bieniek A, Szepietowski J, Piotrowska-Kempisty H, Dzięgiel P, Mozdziak P, Kempisty B. Adipose derived stem cells - Sources, differentiation capacity and a new target for reconstructive and regenerative medicine. Biomed Pharmacother 2025; 186:118036. [PMID: 40194335 DOI: 10.1016/j.biopha.2025.118036] [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: 01/27/2025] [Revised: 03/28/2025] [Accepted: 04/03/2025] [Indexed: 04/09/2025] Open
Abstract
Adipose-derived stem cells (ADSCs) are mesenchymal stem cells (MSCs) derived from adipose tissue with mesenchymal lineage differentiation potential and remarkable potential in regenerative medicine. ADSCs are easily sourced from adipose tissue, share regenerative characteristics akin to other MSCs. Their convenient adherence to plastic culture flasks, coupled with their capacity for in vitro expansion and multi-lineage differentiation, underscores their promise as a robust tool for tissue repair and enhancement. The accessibility of human adipose tissue and the development of minimally invasive isolation protocols have further propelled the autologous use of ADSCs, fueling excitement in both organ repair and regenerative medicine. Consequently, research in ADSCsis experiencing rapid growth. A detailed overview of the current landscape of ADSCs isolation and differentiation capacity including the latest advancements in ADSCs usage, encompassing ongoing clinical investigations are important considerations to understand their potential to shape the landscape of regenerative medicine.
Collapse
Affiliation(s)
- Małgorzata Suchanecka
- Division of Anatomy, Department of Human Morphology and Embryology, Faculty of Medicine, Wroclaw Medical University, Wroclaw 50-368, Poland
| | - Joanna Grzelak
- Division of Anatomy, Department of Human Morphology and Embryology, Faculty of Medicine, Wroclaw Medical University, Wroclaw 50-368, Poland
| | - Maryam Farzaneh
- Fertility, Infertility and Perinatology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Shirin Azizidoost
- Atherosclerosis Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mahrokh Abouali Gale Dari
- Department of Obstetrics and Gynecology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Małgorzata Józkowiak
- Division of Anatomy, Department of Human Morphology and Embryology, Faculty of Medicine, Wroclaw Medical University, Wroclaw 50-368, Poland; Department of Toxicology, Poznan University of Medical Sciences, Poznań 61-631, Poland
| | - Krzysztof Data
- Division of Anatomy, Department of Human Morphology and Embryology, Faculty of Medicine, Wroclaw Medical University, Wroclaw 50-368, Poland
| | - Dominika Domagała
- Division of Anatomy, Department of Human Morphology and Embryology, Faculty of Medicine, Wroclaw Medical University, Wroclaw 50-368, Poland
| | - Julia Niebora
- Division of Anatomy, Department of Human Morphology and Embryology, Faculty of Medicine, Wroclaw Medical University, Wroclaw 50-368, Poland
| | - Katarzyna Kotrych
- Department of General and Dental Radiology, Pomeranian Medical University in Szczecin, Powstańców Wielkopolskich 72, Szczecin 70-111, Poland
| | - Bogusław Czerny
- Department of General Pharmacology and Pharmacoeconomics, Pomeranian Medical University in Szczecin, Żołnierska 48, Szczecin 71-230, Poland; Department of Stem Cells and Regenerative Medicine, Institute of Natural Fibres and Medicinal Plants, Kolejowa 2, Plewiska 62-064, Poland
| | - Adam Kamiński
- Department of Pediatric Orthopedics and Musculosceletal Oncology, Pomeranian Medical University
| | | | - Andrzej Bieniek
- University Center for General and Oncological Dermatology, Wroclaw Medical University, Wroclaw 50-367, Poland
| | - Jacek Szepietowski
- Faculty of Medicine, Wroclaw University of Science and Technology, Wroclaw, Poland; Department of Dermato-Venereology, 4th Military Hospital, Wroclaw, Poland
| | - Hanna Piotrowska-Kempisty
- Department of Toxicology, Poznan University of Medical Sciences, Poznań 61-631, Poland; Department of Basic and Preclinical Sciences, Institute of Veterinary Medicine, Nicolaus Copernicus University in Torun, Torun 87-100, Poland
| | - Piotr Dzięgiel
- Division of Histology and Embryology, Department of Human Morphology and Embryology, Wroclaw Medical University, Wroclaw 50-368, Poland
| | - Paul Mozdziak
- Graduate Physiology Program, North Carolina State University, Raleigh, NC 27695, USA
| | - Bartosz Kempisty
- Division of Anatomy, Department of Human Morphology and Embryology, Faculty of Medicine, Wroclaw Medical University, Wroclaw 50-368, Poland; Center of Assisted Reproduction, Department of Obstetrics and Gynecology, University Hospital and Masaryk University, Brno 625 00, Czech Republic; Department of Veterinary Surgery, Institute of Veterinary Medicine, Nicolaus Copernicus University in Torun, Torun 87-100, Poland; North Carolina State University College of Agriculture and Life Sciences, Raleigh, NC 27695, USA.
| |
Collapse
|
3
|
Xu F, Zhang Q, Liu Y, Tang R, Li H, Yang H, Lin L. The role of exosomes derived from various sources in facilitating the healing of chronic refractory wounds. Pharmacol Res 2025; 216:107753. [PMID: 40311956 DOI: 10.1016/j.phrs.2025.107753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2025] [Revised: 04/23/2025] [Accepted: 04/27/2025] [Indexed: 05/03/2025]
Abstract
Chronic refractory wounds (CRWs) represent a common and challenging issue in clinical practice, including diabetic foot ulcers, pressure ulcers, venous ulcers, and arterial ulcers. These wounds significantly impact patients' quality of life and may lead to severe consequences such as amputation. Their treatment requires a comprehensive consideration of both the patient's overall physical condition and the local wound situation. The major challenges in treatment include complex pathogenesis, a long treatment cycle, a high recurrence rate, and heavy economic pressure on the patients. Exosomes represent an emerging therapeutic modality with characteristics such as low immunogenicity, good biostability, and high targeting efficiency in the treatment of diseases. Exosomes derived from different sources exhibit heterogeneity, demonstrating their respective advantages and unique properties in treatment. This article delves into the potential applications and mechanisms of action of exosomes from various sources in the treatment of CRWs, aiming to provide new perspectives and ideas for the management of such wounds.
Collapse
Affiliation(s)
- Fengdan Xu
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs,Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Qiling Zhang
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs,Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Yuling Liu
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs,Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Ruying Tang
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs,Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Hui Li
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs,Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China; Institute of Traditional Chinese Medicine Health Industry, China Academy of Chinese Medical Sciences, Nanchang 330000, China.
| | - Hongjun Yang
- China Academy of Chinese Medical Sciences, Beijing 100700, China.
| | - Longfei Lin
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs,Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China.
| |
Collapse
|
4
|
Yu H, Wang B, Li Z, Liu K, Chen W, Zhao S, Zhou Y, Wang G, Zhou Y, Chen Y, Chen H, Lai Y, Wang Q, Wang J, Ni B, Zhang D, Pan C, He Y, Li L. Tβ4-exosome-loaded hemostatic and antibacterial hydrogel to improve vascular regeneration and modulate macrophage polarization for diabetic wound treatment. Mater Today Bio 2025; 31:101585. [PMID: 40070869 PMCID: PMC11893380 DOI: 10.1016/j.mtbio.2025.101585] [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: 12/25/2024] [Revised: 01/29/2025] [Accepted: 02/16/2025] [Indexed: 03/14/2025] Open
Abstract
Diabetic wounds often exhibit delayed healing due to compromised vascular function and intensified inflammation. In this study, we overexpressed Thymosin β4 (Tβ4) in Adipose-Derived Stem Cells (ADSCs) to produce Exosomes (Exos) rich in Tβ4. We then utilized a dual photopolymerizable hydrogel composed of Hyaluronic Acid Methacryloyl (HAMA) and Poly-L-lysine Methacryloyl (PLMA) for the sustained release of Tβ4-Exos on diabetic wounds. The results showed that Tβ4-Exos could stimulate angiogenesis and collagen synthesis, and mitigate inflammation in diabetic wounds by promoting the polarization of M1-type macrophages and inhibiting that of M2-type macrophages. Furthermore, Tβ4-Exos was found to activate the PI3K/AKT/mTOR/HIF-1a signaling pathway, thereby enhancing vascular proliferation. In summary, the sustained release of Tβ4-Exos in HAMA-PLMA (HP) hydrogel and the management of inflammation through the upregulation of the HIF-1a pathway and modulation of macrophage polarization in vascular proliferation significantly accelerated the healing process of diabetic wounds.
Collapse
Affiliation(s)
- Hua Yu
- Department of Plastic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Bin Wang
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Zihao Li
- Department of Plastic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Kaibo Liu
- Department of Plastic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Wanying Chen
- Department of Plastic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Songyun Zhao
- Department of Plastic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yu Zhou
- Department of Plastic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Gaoyi Wang
- Department of Plastic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yaqin Zhou
- Department of Plastic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yanming Chen
- Department of Plastic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Housheng Chen
- Department of Plastic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yunning Lai
- Department of Plastic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Quan Wang
- Department of Plastic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jingping Wang
- Department of Plastic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Binting Ni
- Department of Plastic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Dupiao Zhang
- Department of Wound Healing, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Chuanmeng Pan
- Department of Plastic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yucang He
- Department of Plastic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Liqun Li
- Department of Plastic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- National Key Clinical Specialty (Wound Healing), The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
5
|
Wang C, Gao G, Che Q, Zheng S, Yang Y, Li T, Zhai X, Lu Y, Huang B, Yu T, Zhao K, Liu M. Deciphering the value of anoikis-related genes in prognosis, immune microenvironment, and drug sensitivity of laryngeal squamous cell carcinoma. Pathol Res Pract 2025; 268:155849. [PMID: 40020330 DOI: 10.1016/j.prp.2025.155849] [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: 11/01/2024] [Revised: 01/20/2025] [Accepted: 02/20/2025] [Indexed: 03/03/2025]
Abstract
BACKGROUND Laryngeal squamous cell carcinoma (LSCC) is a prevalent malignancy characterized by high metastatic potential and poor prognosis. Anoikis, an apoptotic pathway triggered by detachment from the extracellular matrix (ECM), acts as a barrier against cancer metastasis, so it is necessary to explore the role of anoikis-related genes (ARGs) in LSCC. METHODS Multivariate Cox regression analysis was used to construct prognostic model. A nomogram integrating risk scores with clinicopathological characteristics was constructed for prognosis. Spearman correlation analysis linked ARGs to the tumor microenvironment (TME) and immune infiltration. We also predicted IC50 values for various chemotherapeutic agents by risk group and selected three drugs (LGK974, OSI-027, and OF-1) for molecular docking with MMP3. TCGA datasets was used to evaluate the expression profile of MMP3 and TIMP1 in LSCC. In vitro assays were conducted to confirm the function of target gene in LSCC. RESULTS We identified 19 ARGs associated with LSCC prognosis and developed a prognostic model, which subsequently classified patients into high- and low-risk groups based on median risk scores. Nomogram we established demonstrated excellent predictive performance. Low-risk individuals exhibited significantly higher immunophenotype (IPS) scores and elevated levels of immune cell components than high-risk counterparts (p < 0.05). MMP3 demonstrating strong binding affinity with selected drugs. Analysis of TCGA datasets revealed higher TIMP1 and MMP3 expression in LSCC tissues. CONCLUSIONS Our prognostic signature effectively predicts LSCC prognosis, with MMP3 identified as a potential novel biomarker for LSCC treatment. Furthermore, our findings underscore the critical role of immune-based therapies in improving outcomes, especially for low-risk patients.
Collapse
Affiliation(s)
- Cheng Wang
- Department of Otolaryngology Head and Neck Surgery, Hainan Hospital of Chinese PLA General Hospital, Sanya 572013, China
| | - Ge Gao
- Medical School of Chinese PLA, Beijing 100853, China
| | - Qin Che
- Department of Otolaryngology Head and Neck Surgery, Hainan Hospital of Chinese PLA General Hospital, Sanya 572013, China
| | - Shikang Zheng
- Department of Otolaryngology Head and Neck Surgery, Hainan Hospital of Chinese PLA General Hospital, Sanya 572013, China
| | - Ying Yang
- Department of Otolaryngology Head and Neck Surgery, Hainan Hospital of Chinese PLA General Hospital, Sanya 572013, China
| | - Tian Li
- Tianjin Medical University, Tianjin 300100, China
| | - Xingyou Zhai
- Department of Otolaryngology Head and Neck Surgery, Hainan Hospital of Chinese PLA General Hospital, Sanya 572013, China
| | - Yuehao Lu
- School of Clinical Medicine, Shandong Second Medical University, Weifang 261042, China
| | - Bangqing Huang
- Department of Otolaryngology Head and Neck Surgery, Hainan Hospital of Chinese PLA General Hospital, Sanya 572013, China
| | - Ting Yu
- Department of Otolaryngology Head and Neck Surgery, Hainan Hospital of Chinese PLA General Hospital, Sanya 572013, China
| | - Kai Zhao
- Department of Otolaryngology Head and Neck Surgery, Hainan Hospital of Chinese PLA General Hospital, Sanya 572013, China.
| | - Mingbo Liu
- Department of Otolaryngology Head and Neck Surgery, Hainan Hospital of Chinese PLA General Hospital, Sanya 572013, China; Department of Otolaryngology Head and Neck Surgery, Chinese PLA General Hospital, Beijing 100853, China.
| |
Collapse
|
6
|
Dong Z, Li S, Huang Y, Chen T, Ding Y, Tan Q. RNA N 6-methyladenosine demethylase FTO promotes diabetic wound healing through TRIB3-mediated autophagy in an m 6A-YTHDF2-dependent manner. Cell Death Dis 2025; 16:222. [PMID: 40157922 PMCID: PMC11954964 DOI: 10.1038/s41419-025-07494-3] [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: 10/04/2024] [Revised: 02/01/2025] [Accepted: 02/27/2025] [Indexed: 04/01/2025]
Abstract
N6-methyladenosine (m6A) RNA modification impaired autophagy results in delayed diabetic wound healing. In this study, it was found that fat mass and obesity-associated protein (FTO) was significantly downregulated in the epidermis of diabetic patients, STZ-induced mice and db/db mice (type I and II diabetic mice) with prolonged hyperglycemia, as well as in different types of keratinocyte cell lines treated with short-term high glucose medium. The knockout of FTO affected the biological functions of keratinocytes, including enhanced apoptosis, inhibited autophagy, and delayed wound healing, producing consistent results with high-glucose medium treatment. High-throughput analysis revealed that tribbles pseudokinase 3 (TRIB3) served as the downstream target gene of FTO. In addition, both in vitro and in vivo experiments, TRIB3 overexpression partially rescued biological functions caused by FTO-depletion, promoting keratinocyte migration and proliferation via autophagy. Epigenetically, FTO modulated m6A modification in the 3'UTR of TRIB3 mRNA and enhanced TRIB3 stability in a YTHDF2-dependent manner. Collectively, this study identifies FTO as an accelerator of diabetic wound healing and modulates autophagy via regulating TRIB3 in keratinocytes, thereby benefiting the development of a m6A-targeted therapy for refractory diabetic wounds.
Collapse
Affiliation(s)
- Zheng Dong
- Department of Burn and Plastic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, 210008, China
| | - Shiyan Li
- Department of Burn and Plastic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, 210008, China
| | - Yumeng Huang
- Department of Burns and Plastic Surgery, Nanjing Drum Tower Hospital Clinical College of Jiangsu University, Nanjing, Jiangsu, 210008, China
| | - Tianzhe Chen
- Department of Burn and Plastic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, 210008, China
| | - Youjun Ding
- Department of Burns and Plastic Surgery, Nanjing Drum Tower Hospital Clinical College of Jiangsu University, Nanjing, Jiangsu, 210008, China
| | - Qian Tan
- Department of Burn and Plastic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, 210008, China.
| |
Collapse
|
7
|
Zhu Y, Yang H, Xue Z, Tang H, Chen X, Liao Y. Mesenchymal stem cells-derived small extracellular vesicles and apoptotic extracellular vesicles for wound healing and skin regeneration: a systematic review and meta-analysis of preclinical studies. J Transl Med 2025; 23:364. [PMID: 40128791 PMCID: PMC11934660 DOI: 10.1186/s12967-024-05744-0] [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: 07/06/2024] [Accepted: 10/07/2024] [Indexed: 03/26/2025] Open
Abstract
BACKGROUND Studies examining the therapeutic potential of Mesenchymal stem cells-derived extracellular vesicles (MSC-EVs) in wound healing and skin regeneration have progressed rapidly. Prior to considering clinical translation, a systematic and comprehensive understanding of these experimental details and the overall impact of MSC-EVs on skin regeneration is necessary. METHODS 83 studies were identified in Web of Science, Embase, and PubMed that satisfied a set of prespecified inclusion criteria. A random effects meta-analysis was conducted for wound closure rate, scar width, blood vessel density and collagen deposition. CONCLUSIONS Our findings demonstrate clear potential of MSC-EVs to be developed as therapy for wound healing and skin regeneration both in diabetic and non-diabetic animal models. Moreover, subgroup analyses demonstrated that apoptotic small extracellular vesicles (ApoSEVs) showed better efficacy than apoptotic bodies (ApoBDs) and small extracellular vesicles (sEVs) in wound closure outcome and collagen deposition, while sEVs displayed better than ApoEVs in revascularization. Among frequently used routes of administration, subcutaneous injection displayed a greater improvement to wound closure, collagen deposition and revascularization as compared to dressing/covering. Among easier-access source of MSCs, ADSCs demonstrated the best effect in wound closure rate and collagen deposition, as compared, BMMSCs displayed better in revascularization. Additionally, high heterogeneity observed in collection conditions, separation methods, storage methods, modifications, treatment dose, administration route, and frequency of MSC-EVs underscores the urgent need for standardization in these areas, prior to clinical translation. PROTOCOL REGISTRATION PROSPERO CRD42024499172.
Collapse
Affiliation(s)
- Yufan Zhu
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, Guangdong, 510515, P. R. China
| | - Han Yang
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, Guangdong, 510515, P. R. China
| | - Zhixin Xue
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, Guangdong, 510515, P. R. China
| | - Haojing Tang
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, Guangdong, 510515, P. R. China
| | - Xihang Chen
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, Guangdong, 510515, P. R. China.
| | - Yunjun Liao
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, Guangdong, 510515, P. R. China.
| |
Collapse
|
8
|
Wu D, Liu R, Cen X, Dong W, Chen Q, Lin J, Wang X, Ling Y, Mao R, Sun H, Huang R, Su H, Xu H, Qin D. Preclinical study of engineering MSCs promoting diabetic wound healing and other inflammatory diseases through M2 polarization. Stem Cell Res Ther 2025; 16:113. [PMID: 40038782 PMCID: PMC11881511 DOI: 10.1186/s13287-025-04248-y] [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: 08/29/2024] [Accepted: 02/20/2025] [Indexed: 03/06/2025] Open
Abstract
BACKGROUND Diabetic foot ulcer (DFU) represents a common and severe complication of diabetes mellitus. Effective and safe treatments need to be developed. Mesenchymal stem cells (MSCs) have demonstrated crucial roles in tissue regeneration, wound repair and inflammation regulation. However, the function is limited. The safety and efficacy of gene-modified MSCs is unknown. Therefore, this study aimed to investigate whether genetically modified MSCs with highly efficient expression of anti-inflammatory factors promote diabetic wound repair by regulating macrophage phenotype transition. This may provide a new approach to treating diabetic wound healing. METHODS In this study, human umbilical cord-derived MSCs (hUMSCs) were genetically modified using recombinant lentiviral vectors to simultaneously overexpress three anti-inflammatory factors, interleukin (IL)-4, IL-10, IL-13 (MSCs-3IL). Cell counting kit-8, flow cytometry and differentiation assay were used to detect the criteria of MSCs. Overexpression efficiency was evaluated using flow cytometry, quantitative real-time PCR, Western blot, enzyme-linked immunosorbent assay, and cell scratch assay. We also assessed MSCs-3IL's ability to modulate Raw264.7 macrophage phenotype using flow cytometry and quantitative real-time PCR. In addition, we evaluated diabetic wound healing through healing rate calculation, HE staining, Masson staining, and immunohistochemical analysis of PCNA, F4/80, CD31, CD86, CD206, IL-4, IL-10 and IL-13. In addition, we evaluated the safety of the MSCs-3IL cells and the effect of the cells on several other models of inflammation. RESULTS MSCs-3IL efficiently expressed high levels of IL-4 and IL-10 (mRNA transcription increased by 15,000-fold and 800,000-fold, protein secretion 400 and 200 ng/mL), and IL-13 (mRNA transcription increased by 950,000-fold, protein secretion 6 ng/mL). MSCs-3IL effectively induced phenotypic polarization of pro-inflammatory M1-like macrophages (M1) towards anti-inflammatory M2-like macrophages (M2). The enhancement of function does not change the cell phenotype. The dynamic distribution in vivo was normal and no karyotype variation and tumor risk was observed. In a mouse diabetic wound model, MSCs-3IL promoted diabetic wound healing with a wound closure rate exceeding 96% after 14 days of cell treatment. The healing process was aided by altering macrophage phenotype (reduced CD86 and increased CD206 expression) and accelerating re-epithelialization. CONCLUSIONS In summary, our study demonstrates that genetically modified hUMSCs effectively overexpressed three key anti-inflammatory factors (IL-4, IL-10, IL-13). MSCs-3IL-based therapy enhances diabetic wound healing with high efficiency and safety. This suggests that genetically modified hUMSCs could be used as a novel therapeutic approach for DFU repair.
Collapse
Affiliation(s)
- Di Wu
- Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Rencun Liu
- Shandong Province Key Laboratory of Detection Technology for Tumour Makers, School of Chemistry and Chemical Engineering, Linyi University, Linyi, China
| | - Xiaotong Cen
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, China
| | - Wanwen Dong
- Bioland Laboratory, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China
| | - Qing Chen
- Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Jiali Lin
- Bioland Laboratory, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China
| | - Xia Wang
- Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Yixia Ling
- Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Rui Mao
- Laboratory Animal Research Center, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Haitao Sun
- Neurosurgery Centre, Department of Cerebrovascular Surgery, Engineering Technology Research Centre of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, The National Key Clinical Specialty, Southern Medical University, Guangzhou, Guangdong, China
| | - Rui Huang
- The Fifth Clinical College, Guangzhou Medical University, Guangzhou, China
| | - Huanxing Su
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China.
- , No.621 Gangwan Road, Huangpu District, Guangzhou, China.
| | - Hongjie Xu
- Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China.
- , No.621 Gangwan Road, Huangpu District, Guangzhou, China.
| | - Dajiang Qin
- Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China.
- Bioland Laboratory, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China.
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong SAR, China.
- , No.621 Gangwan Road, Huangpu District, Guangzhou, China.
| |
Collapse
|
9
|
Gowtham A, Kaundal RK. Exploring the ncRNA landscape in exosomes: Insights into wound healing mechanisms and therapeutic applications. Int J Biol Macromol 2025; 292:139206. [PMID: 39732230 DOI: 10.1016/j.ijbiomac.2024.139206] [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: 10/26/2024] [Revised: 12/16/2024] [Accepted: 12/24/2024] [Indexed: 12/30/2024]
Abstract
Exosomal non-coding RNAs (ncRNAs), including miRNAs, lncRNAs, and circRNAs, have emerged as crucial modulators in cellular signaling, influencing wound healing processes. Stem cell-derived exosomes, which serve as vehicles for these ncRNAs, show remarkable therapeutic potential due to their ability to modulate wound healing stages, from initial inflammation to collagen formation. These ncRNAs act as molecular signals, regulating gene expression and protein synthesis necessary for cellular responses in healing. Wound healing is a complex, staged process involving inflammation, hemostasis, fibroblast proliferation, angiogenesis, and tissue remodeling. Stem cell-derived exosomal ncRNAs enhance these stages by reducing excessive inflammation, promoting anti-inflammatory responses, guiding fibroblast and keratinocyte maturation, enhancing vascularization, and ensuring organized collagen deposition. Their molecular cargo, particularly ncRNAs, specifically targets pathways to aid chronic wound repair and support scarless regeneration. This review delves into the unique composition and signaling roles of Stem cell-derived exosomes and ncRNAs, highlighting their impact across wound healing stages and their potential as innovative therapeutics. Understanding the interaction between exosomal ncRNAs and cellular signaling pathways opens new avenues in regenerative medicine, positioning Stem cell-derived exosomes and their ncRNAs as promising molecular-level interventions in wound healing.
Collapse
Affiliation(s)
- A Gowtham
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Raebareli (NIPER-R), Transit Campus, Bijnor-Sisendi Road, Sarojini Nagar, Near CRPF Base Camp, Lucknow, UP 226002, India
| | - Ravinder K Kaundal
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Raebareli (NIPER-R), Transit Campus, Bijnor-Sisendi Road, Sarojini Nagar, Near CRPF Base Camp, Lucknow, UP 226002, India.
| |
Collapse
|
10
|
Yang P, Lu Y, Gou W, Qin Y, Zhang X, Li J, Zhang Q, Zhang X, He D, Wang Y, Xue D, Liu M, Chen Y, Zhou J, Zhang X, Lv J, Tan J, Luo G, Zhang Q. Andrias davidianus Derived Glycosaminoglycans Direct Diabetic Wound Repair by Reprogramming Reparative Macrophage Glucolipid Metabolism. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2417801. [PMID: 39967388 DOI: 10.1002/adma.202417801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Revised: 01/26/2025] [Indexed: 02/20/2025]
Abstract
Harnessing cross-species regenerative cues to direct human regenerative potential is increasingly recognized as an excellent strategy in regenerative medicine, particularly for addressing the challenges of impaired wound healing in aging populations. The skin mucus of Andrias davidianus plays a critical role in self-protection and tissue repair, yet the fundamental regenerative factors and mechanisms involved remain elusive. Here, this work presents evidence that glycosaminoglycans (GAGs) derived from the skin secretion of Andrias davidianus (SAGs) serve as potent mediators of angiogenesis and inflammatory remodeling, facilitating efficient healing of diabetic wounds. Mechanistic studies reveal that SAGs promote macrophage polarization toward an anti-inflammatory and pro-regenerative phenotype (CD206+/Arg1+) via glucolipid metabolic reprogramming. This process suppresses excessive inflammation and enhances the expression of VEGF and IL-10 to create a facilitative microenvironment for tissue regeneration. Additionally, this work develops SAGs-GelMA composite microspheres that address multiple stages of wound healing, including rapid hemostasis, exudate control, and activation of endogenous regenerative processes. This engineered approach significantly improves the scarless healing of diabetic wounds by facilitating the recruitment and activation of reparative macrophages. The findings offer new insights into the regenerative mechanisms of Andrias davidianus and highlight the potential therapeutic application of SAGs in tissue repair.
Collapse
Affiliation(s)
- Peng Yang
- Institute of Burn Research, State Key Laboratory of Trauma and Chemical Poisoning, Southwest Hospital, Third Military Medical University, Chongqing, 400038, China
| | - Yifei Lu
- Institute of Burn Research, State Key Laboratory of Trauma and Chemical Poisoning, Southwest Hospital, Third Military Medical University, Chongqing, 400038, China
| | - Weiming Gou
- Institute of Burn Research, State Key Laboratory of Trauma and Chemical Poisoning, Southwest Hospital, Third Military Medical University, Chongqing, 400038, China
| | - Yiming Qin
- Department of Dermatology and Laboratory of Dermatology, Clinical Institute of Inflammation and Immunology, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xingyue Zhang
- Department of Dermatology and Laboratory of Dermatology, Clinical Institute of Inflammation and Immunology, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jingyuan Li
- Department of Dermatology, The Seventh Medical Center of Chinese PLA General Hospital, Beijing, 100700, China
| | - Qiong Zhang
- Institute of Burn Research, State Key Laboratory of Trauma and Chemical Poisoning, Southwest Hospital, Third Military Medical University, Chongqing, 400038, China
| | - Xiaorong Zhang
- Institute of Burn Research, State Key Laboratory of Trauma and Chemical Poisoning, Southwest Hospital, Third Military Medical University, Chongqing, 400038, China
- Department of Pathology, Southwest Hospital, Third Military Medical University, Chongqing, 400038, China
| | - Dengfeng He
- Institute of Burn Research, State Key Laboratory of Trauma and Chemical Poisoning, Southwest Hospital, Third Military Medical University, Chongqing, 400038, China
| | - Yangping Wang
- Institute of Burn Research, State Key Laboratory of Trauma and Chemical Poisoning, Southwest Hospital, Third Military Medical University, Chongqing, 400038, China
| | - Dongdong Xue
- Institute of Burn Research, State Key Laboratory of Trauma and Chemical Poisoning, Southwest Hospital, Third Military Medical University, Chongqing, 400038, China
| | - Menglong Liu
- Institute of Burn Research, State Key Laboratory of Trauma and Chemical Poisoning, Southwest Hospital, Third Military Medical University, Chongqing, 400038, China
| | - Yu Chen
- Institute of Burn Research, State Key Laboratory of Trauma and Chemical Poisoning, Southwest Hospital, Third Military Medical University, Chongqing, 400038, China
| | - Junyi Zhou
- Institute of Burn Research, State Key Laboratory of Trauma and Chemical Poisoning, Southwest Hospital, Third Military Medical University, Chongqing, 400038, China
| | - Ximu Zhang
- Chongqing Key Laboratory of Oral Disease and Biomedical Sciences and Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Stomatological Hospital of Chongqing Medical University, Chongqing, 401174, China
| | - Junjiang Lv
- Chongqing Engineering Research Center of Pharmaceutical Sciences, Chongqing Medical and Pharmaceutical College, Chongqing, 401331, China
| | - Jianglin Tan
- Institute of Burn Research, State Key Laboratory of Trauma and Chemical Poisoning, Southwest Hospital, Third Military Medical University, Chongqing, 400038, China
| | - Gaoxing Luo
- Institute of Burn Research, State Key Laboratory of Trauma and Chemical Poisoning, Southwest Hospital, Third Military Medical University, Chongqing, 400038, China
| | - Qing Zhang
- Institute of Burn Research, State Key Laboratory of Trauma and Chemical Poisoning, Southwest Hospital, Third Military Medical University, Chongqing, 400038, China
| |
Collapse
|
11
|
Chen G, Pei Y, Ye Q, Xie Z, Gyawali L, Liang X. NEDD4L-mediated RASGRP2 suppresses high-glucose and oxLDL-induced vascular endothelial cell dysfunctions by activating Rap1 and R-Ras. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2024; 1871:119844. [PMID: 39260747 DOI: 10.1016/j.bbamcr.2024.119844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 08/30/2024] [Accepted: 08/31/2024] [Indexed: 09/13/2024]
Abstract
BACKGROUND Ras guanyl-releasing protein 2 (RASGRP2) is an important regulator mediating endothelial cell function. However, whether RASGRP2 mediates diabetes mellitus (DM)-related atherosclerosis (AS) progression by regulating endothelial cell functions is unknown. METHODS Human cardiac microvascular endothelial cells (HCMECs) were treated with high-glucose (HG) and oxidized low-density lipoprotein (oxLDL). The expression of RASGRP2 and neural precursor cell expressed developmentally downregulated 4-like (NEDD4L) was examined by quantitative real-time PCR and western blot (WB). Cell viability, apoptosis, migration, angiogenesis were detected by CCK8 assay, flow cytometry, transwell assay and tube formation assay. ROS production and cell permeability were tested to assess cell function. Rap1 and R-Ras protein levels were examined using WB. The interaction between RASGRP2 and NEDD4L was confirmed by Co-IP assay and ubiquitination assay. Exosomes were isolated from adipose-derived MSC (ADMSC)-transfected RASGRP2 overexpression vector, and then co-cultured with HG + oxLDL-induced HCMECs. RESULTS RASGRP2 was lowly expressed in HG + oxLDL-induced HCMECs. RASGRP2 overexpression inhibited HG + oxLDL-induced HCMECs permeability, apoptosis and ROS production, while accelerated cell viability, migration and angiogenesis. NEDD4L could interact with RASGRP2 by ubiquitination, thus inhibiting RASGRP2 protein stability to degrade its expression. Functional experiments showed that NEDD4L knockdown suppressed HG + oxLDL-induced HCMECs dysfunction, while these effects were reversed by RASGRP2 downregulation. ADMSC-Exo overexpressed RASGRP2 could promote cell viability, migration and angiogenesis, while suppress permeability, apoptosis and ROS production in HG + oxLDL-induced HCMECs. CONCLUSION Our data showed that targeting NEDD4L/RASGRP2 axis or inducing RASGRP2-modified ADMSC-Exo might be the efficient strategy for alleviating DM-related AS.
Collapse
Affiliation(s)
- Guozhu Chen
- The Second Affiliated Hospital of Chongqing Medical University Cardiology Department, Chongqing, China.
| | - Yisong Pei
- The Second Affiliated Hospital of Chongqing Medical University Cardiology Department, Chongqing, China
| | - Qiaoling Ye
- The Second Affiliated Hospital of Chongqing Medical University Cardiology Department, Chongqing, China
| | - Zulong Xie
- The Second Affiliated Hospital of Chongqing Medical University Cardiology Department, Chongqing, China
| | | | - Xing Liang
- The Second Affiliated Hospital of Chongqing Medical University Cardiology Department, Chongqing, China.
| |
Collapse
|
12
|
Sun X, Ding H, Li X, Wu Y, Huang X. Disulfiram-loaded nanovesicles hydrogel promotes healing of diabetic wound. J Transl Med 2024; 22:1066. [PMID: 39593097 PMCID: PMC11600750 DOI: 10.1186/s12967-024-05875-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Accepted: 11/11/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND Traditional methods for treating diabetic wounds are limited in effectiveness because of their long healing times, the risk of immune rejection, and susceptibility to infection. Suppressing neutrophil extracellular traps (NETs) is an effective strategy for reducing persistent inflammation in diabetic wounds. Although disulfiram (DSF) can inhibit the significant increase of NETs in diabetic wounds, oral DSF suffers from rapid and harmful metabolism in the liver. To address these challenges, we developed a nanomedicine formulation in which DSF was incorporated into the hydrogel. METHODS In this study, we developed a DSF-laden sodium alginate hydrogel wound dressing, DEP@SA, and characterized its composition, properties, and performance. We examined the effects of DEP@SA on inflammatory phase-related markers such as NETs and their pathway proteins, inflammatory factors, and macrophage phenotypes in a high-glucose environment in vivo and in vitro. In addition, the effects of DEP@SA on tissue regenerative capacity such as epidermal proliferative migration and angiogenesis, were also assessed. RESULTS The results showed that by utilizing extracellular vesicles as a drug delivery system, we effectively mitigated the degradation of DSF via direct contact with aqueous solutions and ensured the stability of DSF@SA, which could then be applied to diabetic wounds. The inflammatory phase-related indicators revealed that DSF@SA effectively reduced inflammation levels, decreased NETs formation, suppressed the Caspase-1/GSDMD pathway in neutrophils, and promoted the polarization of M2 macrophages. Moreover, the hydrogel accelerated wound healing by promoting angiogenesis and re-epithelialization, thereby shortening the diabetic wound healing time. CONCLUSIONS This study confirmed that the DSF@SA composite dressing has the potential to enhance diabetic wound repair and offers a novel approach for drug reutilization.
Collapse
Affiliation(s)
- Xingzi Sun
- Center for Infection and Immunity and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, 519000, China
| | - Hanxi Ding
- Department of Oncology, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, 519000, China
| | - Xingyu Li
- Center for Infection and Immunity and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, 519000, China
| | - Yongjian Wu
- Center for Infection and Immunity and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, 519000, China.
| | - Xi Huang
- Center for Infection and Immunity and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, 519000, China.
| |
Collapse
|
13
|
Jin C, Zhao R, Hu W, Wu X, Zhou L, Shan L, Wu H. Topical hADSCs-HA Gel Promotes Skin Regeneration and Angiogenesis in Pressure Ulcers by Paracrine Activating PPARβ/δ Pathway. Drug Des Devel Ther 2024; 18:4799-4824. [PMID: 39478872 PMCID: PMC11523932 DOI: 10.2147/dddt.s474628] [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: 05/27/2024] [Accepted: 10/09/2024] [Indexed: 11/02/2024] Open
Abstract
Background Pressure ulcer is common in the bedridden elderly with high mortality and lack of effective treatment. In this study, human-adipose-derived-stem-cells-hyaluronic acid gel (hADSCs-HA gel) was developed and applied topically to treat pressure ulcers, of which efficacy and paracrine mechanisms were investigated through in vivo and in vitro experiments. Methods Pressure ulcers were established on the backs of C57BL/6 mice and treated topically with hADSCs-HA gel, hADSCs, hyaluronic acid, and normal saline respectively. The rate of wound closure was observed continuously during the following 14 days and the wound samples were obtained for Western blot, histopathology, immunohistochemistry, and proteomic analysis. Human dermal fibroblasts (HDFs) and human venous endothelial cells (HUVECs) under normal or hypoxic conditions were treated with conditioned medium of human ADSCs (ADSC-CM), then CCK-8, scratch test, tube formation, and Western blot were conducted to evaluate the paracrine effects of hADSCs and to explore the underlying mechanism. Results The in vivo data demonstrated that hADSCs-HA gel significantly accelerated the healing of pressure ulcers by enhancing collagen expression, angiogenesis, and skin proliferation. The in vitro data revealed that hADSCs strengthened the proliferation and wound healing capabilities of HDFs and HUVECs, meanwhile promoted collagen secretion and tube formation through paracrine mode. ADSC-CM was also proved to exert protective effects on hypoxic HDFs and HUVECs. Besides, the results of proteomic analysis and Western blot elucidated that lipid metabolism and PPARβ/δ pathway mediated the healing effect of hADSCs-HA gel on pressure ulcers. Conclusion Our research showed that topical application of hADSCs-HA gel played an important role in dermal regeneration and angiogenesis. Therefore, hADSCs-HA gel exhibited the potential as a novel stem-cell-based therapeutic strategy of treating pressure ulcers in clinical practices.
Collapse
Affiliation(s)
- Chaoying Jin
- Department of Plastic and Aesthetic Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310003, People’s Republic of China
- School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310020, People’s Republic of China
| | - Ruolin Zhao
- Yichen Biotechnology Co., Ltd, Hangzhou, Zhejiang, 311200, People’s Republic of China
- Fuyang Academy, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 311403, People’s Republic of China
| | - Weihang Hu
- Department of Critical Care Medicine, Zhejiang Hospital, Hangzhou, Zhejiang, 310013, People’s Republic of China
| | - Xiaolong Wu
- Fuyang Academy, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 311403, People’s Republic of China
| | - Li Zhou
- The First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310060, People’s Republic of China
| | - Letian Shan
- Fuyang Academy, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 311403, People’s Republic of China
- The First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310060, People’s Republic of China
| | - Huiling Wu
- Department of Plastic and Aesthetic Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310003, People’s Republic of China
- School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310020, People’s Republic of China
| |
Collapse
|
14
|
Zhu H, Guo X, Zhang Y, Khan A, Pang Y, Song H, Zhao H, Liu Z, Qiao H, Xie J. The Combined Anti-Aging Effect of Hydrolyzed Collagen Oligopeptides and Exosomes Derived from Human Umbilical Cord Mesenchymal Stem Cells on Human Skin Fibroblasts. Molecules 2024; 29:1468. [PMID: 38611748 PMCID: PMC11013016 DOI: 10.3390/molecules29071468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 03/08/2024] [Accepted: 03/18/2024] [Indexed: 04/14/2024] Open
Abstract
Stem cell-derived exosomes (SC-Exos) are used as a source of regenerative medicine, but certain limitations hinder their uses. The effect of hydrolyzed collagen oligopeptides (HCOPs), a functional ingredient of SC-Exos is not widely known to the general public. We herein evaluated the combined anti-aging effects of HCOPs and exosomes derived from human umbilical cord mesenchymal stem cells (HucMSC-Exos) using a senescence model established on human skin fibroblasts (HSFs). This study discovered that cells treated with HucMSC-Exos + HCOPs enhanced their proliferative and migratory capabilities; reduced both reactive oxygen species production and senescence-associated β-galactosidase activity; augmented type I and type III collagen expression; attenuated the expression of matrix-degrading metalloproteinases (MMP-1, MMP-3, and MMP-9), interleukin 1 beta (IL-1β), and tumor necrosis factor-alpha (TNF-α); and decreased the expression of p16, p21, and p53 as compared with the cells treated with HucMSC-Exos or HCOPs alone. These results suggest a possible strategy for enhancing the skin anti-aging ability of HucMSC-Exos with HCOPs.
Collapse
Affiliation(s)
- Huimin Zhu
- Department of Biochemistry and Molecular Biology, Shanxi Key Laboratory of Birth Defect and Cell Regeneration, MOE Key Laboratory of Coal Environmental Pathogenicity and Prevention, Shanxi Medical University, Taiyuan 030001, China; (H.Z.); (X.G.); (Y.Z.); (Y.P.); (H.S.); (H.Z.); (Z.L.)
| | - Xin Guo
- Department of Biochemistry and Molecular Biology, Shanxi Key Laboratory of Birth Defect and Cell Regeneration, MOE Key Laboratory of Coal Environmental Pathogenicity and Prevention, Shanxi Medical University, Taiyuan 030001, China; (H.Z.); (X.G.); (Y.Z.); (Y.P.); (H.S.); (H.Z.); (Z.L.)
| | - Yongqing Zhang
- Department of Biochemistry and Molecular Biology, Shanxi Key Laboratory of Birth Defect and Cell Regeneration, MOE Key Laboratory of Coal Environmental Pathogenicity and Prevention, Shanxi Medical University, Taiyuan 030001, China; (H.Z.); (X.G.); (Y.Z.); (Y.P.); (H.S.); (H.Z.); (Z.L.)
| | - Ajab Khan
- Department of Veterinary Pathology, Faculty of Veterinary and Animal Sciences, The University of Agriculture, Dera Ismail Khan 29050, Pakistan;
| | - Yinuo Pang
- Department of Biochemistry and Molecular Biology, Shanxi Key Laboratory of Birth Defect and Cell Regeneration, MOE Key Laboratory of Coal Environmental Pathogenicity and Prevention, Shanxi Medical University, Taiyuan 030001, China; (H.Z.); (X.G.); (Y.Z.); (Y.P.); (H.S.); (H.Z.); (Z.L.)
| | - Huifang Song
- Department of Biochemistry and Molecular Biology, Shanxi Key Laboratory of Birth Defect and Cell Regeneration, MOE Key Laboratory of Coal Environmental Pathogenicity and Prevention, Shanxi Medical University, Taiyuan 030001, China; (H.Z.); (X.G.); (Y.Z.); (Y.P.); (H.S.); (H.Z.); (Z.L.)
| | - Hong Zhao
- Department of Biochemistry and Molecular Biology, Shanxi Key Laboratory of Birth Defect and Cell Regeneration, MOE Key Laboratory of Coal Environmental Pathogenicity and Prevention, Shanxi Medical University, Taiyuan 030001, China; (H.Z.); (X.G.); (Y.Z.); (Y.P.); (H.S.); (H.Z.); (Z.L.)
| | - Zhizhen Liu
- Department of Biochemistry and Molecular Biology, Shanxi Key Laboratory of Birth Defect and Cell Regeneration, MOE Key Laboratory of Coal Environmental Pathogenicity and Prevention, Shanxi Medical University, Taiyuan 030001, China; (H.Z.); (X.G.); (Y.Z.); (Y.P.); (H.S.); (H.Z.); (Z.L.)
| | - Hua Qiao
- Department of Biochemistry and Molecular Biology, Shanxi Key Laboratory of Birth Defect and Cell Regeneration, MOE Key Laboratory of Coal Environmental Pathogenicity and Prevention, Shanxi Medical University, Taiyuan 030001, China; (H.Z.); (X.G.); (Y.Z.); (Y.P.); (H.S.); (H.Z.); (Z.L.)
| | - Jun Xie
- Department of Biochemistry and Molecular Biology, Shanxi Key Laboratory of Birth Defect and Cell Regeneration, MOE Key Laboratory of Coal Environmental Pathogenicity and Prevention, Shanxi Medical University, Taiyuan 030001, China; (H.Z.); (X.G.); (Y.Z.); (Y.P.); (H.S.); (H.Z.); (Z.L.)
| |
Collapse
|