1
|
Dai X, Dang M, Meng X, Zheng J, Yang Y, Wang L, Wang J, Liang Y, Fei W. Porous Se@SiO 2 nanoparticle composite hydrogels loaded with adipose stem cells improves the local microenvironment to promote rotator cuff tendon-bone healing in rats. J Mater Chem B 2025; 13:5598-5612. [PMID: 40259663 DOI: 10.1039/d4tb02642j] [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: 04/23/2025]
Abstract
Functional repair of the tendon-bone interface poses significant challenges in clinical practice; furthermore, identifying methods to enhance healing at enthesis is a central concern in regenerative medicine. The application of stem cells in the healing process of interface injuries is widespread; however, direct injection of stem cells into this interface leads to significant losses of many stem cells. Oxidative stress significantly influences interface repair, and the role of selenium in mitigating oxidative stress and regulating inflammation has been demonstrated. This study utilised gelatine methacrylate (GelMA) as a stem cell transporter, while porous Se@SiO2 nanoparticles (Se@SiO2 NPs) were incorporated to change the interface microenvironment and facilitate the repair of the tendon-bone interface. Oxidative stress effects were analysed using flow cytometry, immunofluorescence staining, and qRT-PCR. The repair of the enthesis was assessed using histological staining, biomechanical evaluation, and MRI. Se@SiO2 NPs significantly reduced the expression of inflammation-related markers in an in vitro oxidative stress model. Additionally, porous selenium nanocomposite hydrogels loaded with adipose stem cells were implanted into the rat tendon-bone interface. At eight weeks following the procedure, the enthesis exhibited superior collagen fibre continuity and orientation, enhanced bone and fibrocartilage production, and biomechanical functions that were substantially greater than those of the comparison group. This study demonstrates that porous Se@SiO2 NP composite hydrogels with antioxidant and anti-inflammatory properties provide a supportive environment for transplanted stem cells and promote tissue repair.
Collapse
Affiliation(s)
- Xiaomei Dai
- Department of Sports Medicine, Northern Jiangsu People's Hospital, Yangzhou 225001, P. R. China.
| | - Mengbo Dang
- Department of Sports Medicine, Northern Jiangsu People's Hospital, Yangzhou 225001, P. R. China.
| | - Xiangji Meng
- Department of Sports Medicine, Northern Jiangsu People's Hospital Affiliated to Dalian Medical University, Dalian 116000, P. R. China
| | - Jun Zheng
- Department of Sports Medicine, Northern Jiangsu People's Hospital, Yangzhou 225001, P. R. China.
| | - Yuxia Yang
- Department of Sports Medicine, Northern Jiangsu People's Hospital, Yangzhou 225001, P. R. China.
| | - Liang Wang
- Department of Sports Medicine, Northern Jiangsu People's Hospital, Yangzhou 225001, P. R. China.
| | - Jingcheng Wang
- Department of Sports Medicine, Northern Jiangsu People's Hospital, Yangzhou 225001, P. R. China.
| | - Yuan Liang
- Department of Sports Medicine, Northern Jiangsu People's Hospital, Yangzhou 225001, P. R. China.
| | - Wenyong Fei
- Department of Sports Medicine, Northern Jiangsu People's Hospital, Yangzhou 225001, P. R. China.
- Department of Orthopedics, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou 225001, P. R. China
| |
Collapse
|
2
|
Wang H, Gao X, Zhao Y, Sun S, Liu Y, Wang K. Exosome-Loaded GelMA Hydrogel as a Cell-Free Therapeutic Strategy for Hypertrophic Scar Inhibition. Clin Cosmet Investig Dermatol 2025; 18:1137-1149. [PMID: 40351852 PMCID: PMC12065470 DOI: 10.2147/ccid.s520913] [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: 02/05/2025] [Accepted: 04/12/2025] [Indexed: 05/14/2025]
Abstract
Purpose Hypertrophic scar (HS) is a fibrotic proliferative disorder that arises from an abnormal wound healing process. It is a significant clinical challenge, primarily characterized by the excessive accumulation of extracellular matrix (ECM) and abnormal angiogenesis. This study introduces a novel injectable hydrogel system that integrates sustained-release Exosomes for targeted hypertrophic scar modulation. Exosomes (Exos) from adipose-derived stem cells (ASCs) are emerging as promising treatment for hypertrophic scar inhibition. But when treated independently, it must be applied regularly multiple times to maintain its optimal concentration. Gelatin Methacryloyl (GelMA) hydrogel is an ideal biomaterial candidate for engineering skin tissues because of its similarity to ECM, and importantly GelMA hydrogel can maintain drug concentrations via the encapsulation and sustained release of it, which enhances the potential of clinical applications. Methods The Exosome-Loaded GelMA Hydrogel (Exos-GelMA) hydrogel was fabricated and characterized for its pore size and biocompatibility. A rabbit ear HS model was established. Three skin defects on each ear were treated with GelMA hydrogel, Exos-GelMA hydrogel, or left untreated as a blank group. The effects of HS inhibition were assessed through Hematoxylin and Eosin (HE) staining, Masson's trichrome staining, and immunohistochemical staining of Collagen I (COL I), Collagen III (COL III), α-smooth muscle actin (α-SMA), as well as immunofluorescence staining of vascular endothelial growth factor (VEGF). Results The Exos-GelMA hydrogel demonstrated an appropriate pore size distribution, excellent biocompatibility, and enhanced fibroblast proliferation in vitro. In the rabbit ear HS model, the Exos-GelMA hydrogel significantly inhibited excessive collagen fiber deposition and the overexpression of the angiogenic factor VEGF. Quantitative analysis of immunohistochemical and immunofluorescence staining showed comparing to blank group the Exos-GelMA hydrogel significantly reduced COL I deposition by 43%, COL III deposition by 15%, α-SMA expression by 31%, and VEGF expression by 35% at 28 day. Conclusion In summary, the Exos-GelMA composite hydrogel exhibits significant potential for the prevention and treatment of HS. This study supports the feasibility of Exos-GelMA as a cell-free therapeutic approach for the management of HS.
Collapse
Affiliation(s)
- Hui Wang
- School of Nursing, Shandong Second Medical University, Weifang, People’s Republic of China
- Department of Burns and Wound Repair, Weifang People’s Hospital, Shandong Second Medical University, Weifang, People’s Republic of China
| | - Xijuan Gao
- Department of Burns and Wound Repair, Weifang People’s Hospital, Shandong Second Medical University, Weifang, People’s Republic of China
| | - Yanxia Zhao
- Department of Burns and Wound Repair, Weifang People’s Hospital, Shandong Second Medical University, Weifang, People’s Republic of China
| | - Shudong Sun
- Department of Burns and Wound Repair, Weifang People’s Hospital, Shandong Second Medical University, Weifang, People’s Republic of China
| | - Yuxiu Liu
- School of Nursing, Shandong Second Medical University, Weifang, People’s Republic of China
| | - Kun Wang
- Department of Burns and Wound Repair, Weifang People’s Hospital, Shandong Second Medical University, Weifang, People’s Republic of China
| |
Collapse
|
3
|
Li M, Shi H, Dong J, Lu N, Lou J, Xu Y. Mechanisms of Ferroptosis-Related Genes in Gallbladder Cancer Based on Bioinformatics Analysis. Mol Biotechnol 2025; 67:1814-1825. [PMID: 38635107 PMCID: PMC11982096 DOI: 10.1007/s12033-024-01159-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 03/26/2024] [Indexed: 04/19/2024]
Abstract
Gallbladder Cancer (GBC) is a lethal malignancy with limited treatment options and poor prognosis. Recent studies have emphasized the role of ferroptosis, a regulated form of cell death, in various cancers, including GBC. We applied bioinformatics methodologies on four GBC datasets to identify differentially expressed genes (DEGs). An intersection of DEGs from the four datasets with ferroptosis and GBC-associated genes was done to identify key ferroptosis-related genes in GBC. GSVA pathway enrichment analysis and immune cell infiltration assessment were conducted to explore their functional roles and interactions. Seven ferroptosis-related genes, EZH2, MUC1, PVT1, GOT1, CDO1, LIFR, and TFAP2A, were identified to be related to GBC. These genes were associated with vital signaling pathways like the G2/M checkpoint and DNA repair and showed significant correlations with immune cell infiltration in GBC. Receiver Operating Characteristic (ROC) curve analysis revealed their high diagnostic potential, with Area Under the Curve (AUC) values ranging from 0.796 to 0.953. Our findings underscore the pivotal role of ferroptosis in GBC and the potential of ferroptosis-related genes as diagnostic biomarkers. This study lays a foundation for further research into ferroptosis-based therapeutic strategies for GBC.
Collapse
Affiliation(s)
- Miao Li
- Department of Oncology, Ningbo TCM Hospital Affiliated to Zhejiang Chinese Medical University (Ningbo Hospital of Traditional Chinese Medicine), No. 819, Liyuan North Road, Ningbo, 315000, Zhejiang, China.
| | - Hang Shi
- Department of Oncology, Ningbo TCM Hospital Affiliated to Zhejiang Chinese Medical University (Ningbo Hospital of Traditional Chinese Medicine), No. 819, Liyuan North Road, Ningbo, 315000, Zhejiang, China
| | - Jing Dong
- Department of Oncology, Ningbo TCM Hospital Affiliated to Zhejiang Chinese Medical University (Ningbo Hospital of Traditional Chinese Medicine), No. 819, Liyuan North Road, Ningbo, 315000, Zhejiang, China
| | - Ning Lu
- Department of Oncology, Ningbo TCM Hospital Affiliated to Zhejiang Chinese Medical University (Ningbo Hospital of Traditional Chinese Medicine), No. 819, Liyuan North Road, Ningbo, 315000, Zhejiang, China
| | - Jinjie Lou
- Department of Oncology, Ningbo TCM Hospital Affiliated to Zhejiang Chinese Medical University (Ningbo Hospital of Traditional Chinese Medicine), No. 819, Liyuan North Road, Ningbo, 315000, Zhejiang, China
| | - Yangbo Xu
- Department of Oncology, Ningbo TCM Hospital Affiliated to Zhejiang Chinese Medical University (Ningbo Hospital of Traditional Chinese Medicine), No. 819, Liyuan North Road, Ningbo, 315000, Zhejiang, China
| |
Collapse
|
4
|
Wang L, Kang J, Li Y, Xia Y, Li X, Du X, Yin Z, Tian F, Wu F, Zhao B. BMSCs laden gelatin methacrylate (GelMA) hydrogel integrating silk fibroin/hydroxyapatite scaffold with multi-layered-oriented pores for enhanced bone regeneration. Int J Pharm 2025; 675:125495. [PMID: 40154821 DOI: 10.1016/j.ijpharm.2025.125495] [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: 12/21/2024] [Revised: 02/26/2025] [Accepted: 03/16/2025] [Indexed: 04/01/2025]
Abstract
Due to the limited regenerative ability of bone tissue, bone injury repair has always been a complicated problem in clinical treatment. Bone tissue engineering based on cell delivery is an effective method to repair bone defects, but it also puts forward strict requirements on the scaffold used in the repair process and the survival rate of cell inoculation. To address this challenge, we constructed a bone mesenchymal stem cells (BMSCs) laden gelatin methacrylate (GelMA) hydrogel to integrate in silk fibroin (SF) /nano-hydroxyapatite (nHAp) scaffold, building a dual architecture to achieve enhanced angiogenesis and bone regeneration. The GelMA hydrogel prepared by visible photo-crosslinking showed good cell loading capacity, and the multi-layered-oriented pores of the scaffold provided a suitable microenvironment for cell proliferation and nutrient exchange. We further explored the effects of this "dual-system" complex on BMSCs and in a critical-sized rat cranial defect model. The results showed that BMSCs@GelMA-SF/nHAp composite scaffold with directional pore structure was more conducive to the repair of skull defects in rats due to the faster rate of vascularization and osteogenesis, indicating the developed gel-scaffold complex would be a promising therapeutic strategy for the repair of bone defects regeneration.
Collapse
Affiliation(s)
- Lu Wang
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan, Shanxi, China; Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, China
| | - Jie Kang
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan, Shanxi, China; Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, China
| | - Yuanjiao Li
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, China; Academy of Medical Sciences, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yijing Xia
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan, Shanxi, China; Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, China
| | - Xiujuan Li
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan, Shanxi, China; Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, China
| | - Xin Du
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan, Shanxi, China; Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, China
| | - Ziruo Yin
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan, Shanxi, China; Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, China
| | - Feng Tian
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan, Shanxi, China; Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, China
| | - Feng Wu
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan, Shanxi, China; Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, China.
| | - Bin Zhao
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan, Shanxi, China; Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, China.
| |
Collapse
|
5
|
Du T, Zhou L, Liu J, Wang X, Xie H, Yang X, Yang Y. Effectiveness of Microwave Therapy Combined with Berberine /GelMA via COX-2/IL-1β Pathway to Treat Skeletal Muscle Injury: An in vivo Study in Rats. Int J Nanomedicine 2025; 20:5509-5527. [PMID: 40321801 PMCID: PMC12047279 DOI: 10.2147/ijn.s500490] [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: 11/02/2024] [Accepted: 04/03/2025] [Indexed: 05/08/2025] Open
Abstract
Introduction Skeletal muscle injuries are short-term, that occur in people who play sports and train. Regular exercise and sports populations undergo repetitive tearing and regeneration of skeletal muscle, in which muscle damage is a necessary component to produce an oxidative inflammatory response and tissue reconstruction. The primary goals of treating this illness are to reduce the disease process cycle and get rid of symptoms like swelling and inflammation at the site of localized injury. Berberine (BBR) has several pharmacological effects, including anti-inflammatory, anti-tumor, and anti-arrhythmic properties. Methods In order to treat skeletal muscle injuries, a safe and non-toxic nanogel (BBR/GelMA) was developed for efficient berberine delivery. It also investigated whether BBR/GelMA had anti-inflammatory properties via the NF-κB pathway. Microwave irradiation was added to promote the uptake of BBR in BBR/GelMA by injured skeletal muscle and to accelerate the process of injury recovery. Results It turns out that the survival rates of NIH313 and L929 cells decreased to varying degrees in GelMA loaded with different concentrations of BBR, but the survival rates of the two cell lines were the highest at a concentration of 0.125 mg/mL. Conclusion In this experiment, the inhibitory effect of BBR/GelMA on inflammation was studied. After NIH-313 and L929 cells were treated with GelMA loaded with different doses of BBR, it was found that the concentration of BBR/0.5 mg/mL had the best inhibitory effect on these two inflammation-inducing cell lines, and this inhibitory effect was related to the drug loading concentration. On the other hand, BBR/GelMA and microwave therapy can play an anti-inflammatory and repairing role in skeletal muscle through NF-κB pathway. In addition, microwave can accelerate the diffusion of BBR in BBR/GelMA within injured skeletal muscle, speeding up the healing process after skeletal muscle injury and shortening the disease cycle.
Collapse
Affiliation(s)
- Tianhao Du
- Department of Rehabilitation Medicine, General Hospital of Northern Theater Command, Shenyang, People’s Republic of China
- Liaoning University of traditional Chinese Medicine, Shenyang, People’s Republic of China
- Hebei Province Hospital of Traditional Chinese Medicine, Shijiazhuang, People’s Republic of China
| | - Liangliang Zhou
- Department of Rehabilitation Medicine, General Hospital of Northern Theater Command, Shenyang, People’s Republic of China
- Department of Orthopedics, The Fourth Affiliated Hospital of China Medical University, Shenyang, People’s Republic of China
| | - Jia Liu
- Department of Rehabilitation Medicine, General Hospital of Northern Theater Command, Shenyang, People’s Republic of China
| | - Xiao Wang
- Department of Rehabilitation Medicine, General Hospital of Northern Theater Command, Shenyang, People’s Republic of China
- Liaoning University of traditional Chinese Medicine, Shenyang, People’s Republic of China
| | - Haoxu Xie
- Department of Rehabilitation Medicine, General Hospital of Northern Theater Command, Shenyang, People’s Republic of China
- Liaoning University of traditional Chinese Medicine, Shenyang, People’s Republic of China
| | - Xu Yang
- Department of Rehabilitation Medicine, General Hospital of Northern Theater Command, Shenyang, People’s Republic of China
| | - Yingxin Yang
- Department of Rehabilitation Medicine, General Hospital of Northern Theater Command, Shenyang, People’s Republic of China
| |
Collapse
|
6
|
Su Y, Huang Z, Chen Y, Deng J, Huang Y, Xiong W. Exosomes from miR-21-5p-modified adipose-derived stem cells promote wound healing by regulating M2 macrophage polarization in a rodent model of pressure ulcer. J Mol Histol 2025; 56:135. [PMID: 40249566 DOI: 10.1007/s10735-025-10407-5] [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: 02/17/2025] [Accepted: 03/21/2025] [Indexed: 04/19/2025]
Abstract
Pressure ulcers represent a significant healthcare burden worldwide. Numerous research has demonstrated the therapeutic potential of adipose-derived stem cell (ADSC)-derived exosomes in promoting wound healing. This study aims to investigate whether exosomes derived from miRNA-modified ADSCs play a role in pressure ulcers by affecting inflammation and macrophage polarization. ADSCs were identified by detecting the surface markers and multilineage differentiation potential. Lentiviruses carrying miR-21-5p were transduced in ADSCs for stable overexpression. Exosomes were extracted from ADSCs and identified. RT-qPCR was employed to detect RNA levels. A mouse model of pressure ulcers was established, followed by injection of exosomes. DiO staining was conducted to assess exosome biodistribution at wound sites. Hematoxylin-eosin and Masson staining were conducted for histological analysis. Immunofluorescence staining was used to evaluate TNF-α and IL-6 expression in mouse wound tissues. Western blotting was conducted to evaluate protein levels of macrophage polarization markers in vivo and in vitro. The results revealed that exosomes derived from miR-21-5p-overexpressing ADSCs promoted wound healing and reduced inflammatory cytokine expression in mouse wound tissues. Moreover, exosomal miR-21-5p induced macrophage M2 polarization in both mouse wound tissues and bone marrow-derived macrophages. Mechanistically, exosomal miR-21-5p inhibited NF-κB signal transduction in mouse wound tissues. In conclusion, ADSC-derived exosomes promote M2 macrophage polarization and inhibit inflammatory response in pressure ulcers via miR-21-5p delivery.
Collapse
Affiliation(s)
- Yongsheng Su
- Department of Burn and Plastic Surgery, The People's Hospital of Baoan Shenzhen, Shenzhen, 518000, China
| | - Zhibin Huang
- Department of Burn and Plastic Surgery, The People's Hospital of Baoan Shenzhen, Shenzhen, 518000, China
| | - Yuanwen Chen
- Department of Burn and Plastic Surgery, The People's Hospital of Baoan Shenzhen, Shenzhen, 518000, China
| | - Jingcheng Deng
- Department of Burn and Plastic Surgery, The People's Hospital of Baoan Shenzhen, Shenzhen, 518000, China
| | - Yubin Huang
- Department of Burn and Plastic Surgery, The People's Hospital of Baoan Shenzhen, Shenzhen, 518000, China
| | - Wei Xiong
- Department of Burn and Plastic Surgery, Huazhong University of Science and Technology Union Shenzhen Hospital, 89 Taoyuan Road, Nanshan District, Shenzhen, 518000, Guangdong, China.
| |
Collapse
|
7
|
Zhao E, Tang X, Zhao M, Yang L. Biodegradable multifunctional hyaluronic acid hydrogel microneedle band-aids for accelerating skin wound healing. Drug Deliv Transl Res 2025:10.1007/s13346-025-01857-1. [PMID: 40246787 DOI: 10.1007/s13346-025-01857-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/07/2025] [Indexed: 04/19/2025]
Abstract
Wound healing for various diseases and wounds such as diabetes and burns remains a major biomedical challenge. Conventional monotherapy is ineffective, and the efficacy of drug delivery is limited by the depth of drug penetration. In this study, we develop a novel, multifunctional, dissolvable hyaluronic acid (HA) microneedle patch (MN-LTT). Microneedling is biocompatible and delivers the drug in a painless and non-invasive manner. Lidocaine and thrombin are mixed with HA hydrogel and loaded onto the needle tips of the MN-LTT, which facilitates wound repair by continuously delivering the drug deep into the dermis for rapid analgesia and hemostasis. In addition, the backing layer of the MN-LTT is composed of tetracycline hydrochloride and HA hydrogel, and its excellent antimicrobial properties further accelerate wound healing. In a mouse full-thickness skin wound model, MN-LTT accelerated cell proliferation and granulation tissue growth, reduced inflammatory-factor levels, and restored collagen deposition, resulting in complete wound healing within seven days. Thus, the proposed microneedle delivery system achieved rapid hemostatic, analgesic, and bactericidal effects, providing a safer and more effective strategy for wound healing. These features make the multifunctional HA microneedle patch potentially valuable for clinical applications.
Collapse
Affiliation(s)
- Erman Zhao
- Precision Pharmacy & Drug Development Center, Department of Pharmacy, Tangdu Hospital, Air Force Medical University, Xi'an, 710038, China
| | - Xiuling Tang
- Precision Pharmacy & Drug Development Center, Department of Pharmacy, Tangdu Hospital, Air Force Medical University, Xi'an, 710038, China
| | - Minggao Zhao
- Precision Pharmacy & Drug Development Center, Department of Pharmacy, Tangdu Hospital, Air Force Medical University, Xi'an, 710038, China.
| | - Le Yang
- Precision Pharmacy & Drug Development Center, Department of Pharmacy, Tangdu Hospital, Air Force Medical University, Xi'an, 710038, China.
| |
Collapse
|
8
|
Li M, Nie J, Li X, Ye L, Wang Z, Yang J. Exudate Management, Facile Detachment, and Immunometabolism Regulation for Wound Healing Using Breathable Dressings. ACS APPLIED MATERIALS & INTERFACES 2025; 17:22394-22409. [PMID: 40167422 DOI: 10.1021/acsami.5c01729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Developing breathable dressings with multifunctional properties (such as exudate management, easy removal, and immunometabolism regulation) presents significant challenges in wound healing. This study employs the Hofmeister effect to prepare a sodium citrate-cross-linked cryogel (CA-CS) with versatile functions, including porous and loose structures, rapid shape recovery ability, superior fatigue resistance behavior, and outstanding biocompatibility capabilities. The CA-CS cryogels demonstrated strong anti-inflammatory properties by reversing the lipopolysaccharides-induced M1 macrophages and increasing M2 macrophage percentages in vitro. Additionally, these breathable CA-CS cryogels exhibited superior hemostatic activity in vivo. The easily detachable CA-CS cryogels enhanced nutrient exchange, promoted exudate absorption, regulated immune response, and induced metabolic reprogramming, thereby supporting skin regeneration and hair follicle formation in a full-thickness skin defect mouse model. We expect that these CA-CS cryogels will drive the development of next-generation dressings for effective wound regeneration in clinical practice.
Collapse
Affiliation(s)
- Mengxin Li
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
- Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Juan Nie
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Xin Li
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Ling Ye
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
- Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Zhenming Wang
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Jing Yang
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
- Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| |
Collapse
|
9
|
Zare S, Jafarzadeh A, Zare S, Shamloo A. Exploring the dermatological applications of human mesenchymal stem cell secretome: a comprehensive review. Stem Cell Res Ther 2025; 16:177. [PMID: 40221781 PMCID: PMC11993991 DOI: 10.1186/s13287-025-04311-8] [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: 02/12/2025] [Accepted: 04/01/2025] [Indexed: 04/14/2025] Open
Abstract
INTRODUCTION Mesenchymal stem cell (MSC)-derived conditioned media is emerging as a promising alternative to stem cell therapy, owing to its abundant content of growth factors and cytokines. OBJECTIVE This review evaluates the clinical applications of MSC-conditioned media in improving scars, promoting wound healing, stimulating hair growth, and rejuvenating the skin. MATERIALS AND METHODS A thorough search of relevant databases was performed to identify studies meeting the inclusion criteria. From an initial pool of 75 articles, 16 studies published up to 2024 were selected based on their relevance, focus, and alignment with the research objectives. RESULTS Among the 17 selected studies, 5 examined the role of conditioned media in skin rejuvenation, 3 investigated its effects on hair growth, 5 assessed its efficacy in scar treatment, 2 assessed its efficacy in Inflammatory Dermatologic Disease and 2 explored its role in wound healing. All studies reported favorable outcomes, demonstrating significant improvements in scars, hair regrowth, and skin rejuvenation with the application of conditioned media. CONCLUSION This review underscores the potential of MSC-derived conditioned media in dermatology. Several studies also highlighted its enhanced therapeutic effects when combined with adjunctive treatments, such as laser therapy and microneedling, showcasing improved outcomes in dermatological care.
Collapse
Affiliation(s)
- Sona Zare
- Skin and Stem Cell Research Center, Tehran University of Medical Sciences, Tehran, Iran
- Persian Bio-Based Production (PBBP) Company, Sharif University of Technology, Tehran, Iran
- Stem Cell and Regenerative Medicine Institute, Sharif University of Technology, Azadi Street, Tarasht Avenue, Tehran, 1445613131, Iran
- Department of Mechanical Engineering, Sharif University of Technology, Tehran, Iran
| | - Alireza Jafarzadeh
- Department of Dermatology, Rasool Akram Medical Complex Clinical Research Development Center (RCRDC), School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Solmaz Zare
- Persian Bio-Based Production (PBBP) Company, Sharif University of Technology, Tehran, Iran
- Laser Application in Medical Sciences Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amir Shamloo
- Stem Cell and Regenerative Medicine Institute, Sharif University of Technology, Azadi Street, Tarasht Avenue, Tehran, 1445613131, Iran.
- Department of Mechanical Engineering, Sharif University of Technology, Tehran, Iran.
| |
Collapse
|
10
|
Li D, Wang Y, Zhao W, Li L, Zhang P. Gold@Mesoporous Polydopamine Nanocomposite Hydrogel Loaded with Estrogen for the Treatment of Skin Photoaging. Int J Nanomedicine 2025; 20:4571-4587. [PMID: 40242609 PMCID: PMC12002076 DOI: 10.2147/ijn.s511388] [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: 12/10/2024] [Accepted: 04/01/2025] [Indexed: 04/18/2025] Open
Abstract
Introduction Topical application of 17β-estradiol (E2) has been shown to improve various hallmark features of skin aging, including enhancing skin elasticity and hydration, reducing wrinkles, and promoting collagen synthesis. However, the role of estrogen in UVB-induced photoaging of the skin remains unclear. Furthermore, E2's clinical application is limited by issues such as bioavailability and potential adverse effects. Therefore, this study aims to explore the role of E2 in UVB-induced skin photoaging and to prepare a gold (Au)@mesoporous polydopamine (mPDA)-hyaluronic acid (HA)/carboxymethyl chitosan (CMCS) nanoparticle composite hydrogel (Au/E2@mPDA-HCG) for the treatment of skin photoaging. Methods This study successfully fabricated mPDA with a well-defined mesoporous structure and incorporated Au NPs into the mesopores of mPDA using an in situ growth method, thereby constructing Au@mPDA NPs loaded with E2. Subsequently, the Au/E2@mPDA NPs were embedded into a HA/CMCS hydrogel to develop the Au/E2@mPDA-HCG nanoparticle composite hydrogel. The composite hydrogel was characterized through in vitro and in vivo experiments, and its efficacy in improving skin photoaging was evaluated. Results This study revealed that estrogen deficiency significantly exacerbates UVB-induced skin photoaging, likely through mechanisms closely associated with increased oxidative stress and reduced collagen production. Moreover, the Au/E2@mPDA-HCG nanoparticle composite hydrogel demonstrated favorable morphological characteristics and biocompatibility. In vitro and in vivo experimental results indicated that this composite hydrogel effectively enhanced the therapeutic efficacy of E2 in treating skin photoaging, as evidenced by its significant mitigation of oxidative stress and inflammatory responses, along with the promotion of collagen synthesis. Conclusion In conclusion, this study suggests that the combination of E2 with Au@mPDA@HCG nanocomposite hydrogel offers a promising therapeutic strategy for UVB-induced skin photoaging.
Collapse
Affiliation(s)
- Dashuai Li
- Department of Stomatology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, People’s Republic of China
| | - Yonghua Wang
- Department of Ophthalmology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, People’s Republic of China
| | - Wanyi Zhao
- Department of Plastic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, People’s Republic of China
| | - Liqun Li
- Department of Plastic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, People’s Republic of China
| | - Pan Zhang
- Department of Plastic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, People’s Republic of China
| |
Collapse
|
11
|
Li Z, Dong H, Yang S, Wang X, Li Z. An Injectable Ibuprofen Sustained-Release Composite Hydrogel System Effectively Accelerates Diabetic Wound Healing via Anti-Inflammatory Effects and Angiogenesis. Int J Nanomedicine 2025; 20:4535-4550. [PMID: 40236520 PMCID: PMC11998958 DOI: 10.2147/ijn.s504924] [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: 11/06/2024] [Accepted: 03/28/2025] [Indexed: 04/17/2025] Open
Abstract
Purpose Excessive inflammation in diabetic wounds, driven by hyperglycemia, prolongs healing, increases the risk of non-healing ulcers, and can lead to severe complications such as amputation or life-threatening infections. Recurrent wound infections and prolonged treatment impose significant economic and psychological burdens, drastically reducing patients' quality of life. Modulating the inflammatory response is a promising strategy to accelerate diabetic wound healing. Ibuprofen (IBU), a widely used anti-inflammatory and analgesic agent, has the potential to promote healing by mitigating excessive inflammation and alleviating wound-associated pain. However, its clinical application is hindered by poor water solubility and a short half-life. Therefore, a controlled and sustained-release system for IBU could enhance its therapeutic efficacy in diabetic wound management. Materials and Methods Here, we present an in situ multi-crosslinked composite hydrogel system that integrates oxidized alginate (OSA), methacryloylated gelatin (GelMA), and an ibuprofen/amino-modified β-cyclodextrin inclusion complex (IBU/CD-NH2) via ion crosslinking, photocrosslinking, and Schiff-base reactions. Results The optimized hydrogel formulation was synthesized at 35°C, with a P/A molar ratio of 2 and an methacrylamide(MA) volume fraction of 20%. Physicochemical and biocompatibility analyses demonstrated that the IBU-loaded composite hydrogel exhibits enhanced mechanical strength, favorable biocompatibility, tunable degradation, and injectability. This system effectively addresses IBU's solubility and absorption challenges while conforming to wounds of varying shapes and sizes, enabling controlled and sustained drug release. Cellular and animal studies confirmed that the hydrogel continuously and uniformly releases IBU, exerting anti-inflammatory effects while promoting angiogenesis and fibroblast migration. This leads to enhanced granulation tissue formation, collagen deposition, and epidermal regeneration, significantly accelerating wound closure within 14 days. Conclusion By simultaneously suppressing inflammation and stimulating tissue regeneration through controlled IBU release, this hydrogel system offers a highly effective strategy for diabetic wound healing and holds strong potential for clinical application.
Collapse
Affiliation(s)
- Zhibin Li
- Department of Plastic and Aesthetic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, People’s Republic of China
| | - Haijiang Dong
- Department of Plastic and Aesthetic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, People’s Republic of China
| | - Shenyu Yang
- Medical 3D Printing Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, People’s Republic of China
| | - Ximei Wang
- Department of Plastic and Aesthetic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, People’s Republic of China
| | - Zhen Li
- Department of Endovascular Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, People’s Republic of China
| |
Collapse
|
12
|
Wu J, Li S, Wang H, Qi Y, Tao S, Tang P, Liu D. High-yield BMSC-derived exosomes by the 3D culture system to enhance the skin wound repair. Regen Biomater 2025; 12:rbaf022. [PMID: 40309353 PMCID: PMC12041419 DOI: 10.1093/rb/rbaf022] [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: 11/01/2024] [Revised: 03/16/2025] [Accepted: 03/20/2025] [Indexed: 05/02/2025] Open
Abstract
Wound defects pose a substantial challenge in clinical practice, often resulting in prolonged healing times and an elevated risk of infection. Insufficient vascularization is a critical factor that adversely affects wound healing. Exosomes obtained from bone mesenchymal stem cells (BMSC-exos) have demonstrated significant promise in accelerating tissue repair by promoting angiogenesis. However, their limited yield and suboptimal biological functions impede widespread clinical application in enhancing wound healing. Prior research has indicated that 3D cultures can boost exosome secretion when compared to conventional 2D cultures. However, the currently prevalent 3D culture methods often necessitate expensive equipment or cumbersome procedures. This study investigates a cost-effective and user-friendly 3D culture system developed using gelatin methacrylate (GelMA). Our findings indicate that a 5% concentration of GelMA provides an optimal environment for the 3D culture of BMSCs. Furthermore, we observed that 3D culture significantly delays the senescence of BMSCs, thereby creating favorable conditions for the sustained production of exosomes. Additionally, 3D cultivation has the potential to boost exosome secretion and enhance their angiogenic capabilities. In vivo experiments further confirmed that BMSC-exos from a 3D environment exhibit enhanced capabilities to promote wound healing. These results suggest that GelMA-based 3D cultures offer a novel strategy for both industrial production and clinical application of exosomes.
Collapse
Affiliation(s)
- Jie Wu
- Medical School of Chinese PLA, Beijing 100853, China
- Department of Orthopedics, The First Medical Center of Chinese PLA General Hospital, Beijing 100853, China
- Senior Department of Orthopedics, The Fourth Medical Center of PLA General Hospital, Beijing 100142, China
- Department of Orthopedics, The Eighth Medical Center of Chinese PLA General Hospital, Beijing 100091, China
| | - Siqi Li
- Department of Nephrology, The Second Medical Center of Chinese PLA General Hospital, Beijing 100853, China
| | - Hao Wang
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou, Gansu 730030, China
| | - Yuanbo Qi
- Department of Orthopedics, The Eighth Medical Center of Chinese PLA General Hospital, Beijing 100091, China
| | - Sheng Tao
- Senior Department of Orthopedics, The Fourth Medical Center of PLA General Hospital, Beijing 100142, China
- Department of Orthopedics, The Eighth Medical Center of Chinese PLA General Hospital, Beijing 100091, China
| | - Peifu Tang
- Medical School of Chinese PLA, Beijing 100853, China
- Senior Department of Orthopedics, The Fourth Medical Center of PLA General Hospital, Beijing 100142, China
| | - Daohong Liu
- Medical School of Chinese PLA, Beijing 100853, China
- Senior Department of Orthopedics, The Fourth Medical Center of PLA General Hospital, Beijing 100142, China
| |
Collapse
|
13
|
He Y, Lu Y, Li R, Tang Y, Du W, Zhang L, Wu J, Li K, Zhuang W, Lv S, Han Y, Tao B, Deng F, Zhao W, Yu D. CircAars-Engineered ADSCs Facilitate Maxillofacial Bone Defects Repair Via Synergistic Capability of Osteogenic Differentiation, Macrophage Polarization and Angiogenesis. Adv Healthc Mater 2025; 14:e2404501. [PMID: 40035523 PMCID: PMC12004435 DOI: 10.1002/adhm.202404501] [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: 11/13/2024] [Revised: 01/12/2025] [Indexed: 03/05/2025]
Abstract
Adipose-derived stem cells (ADSCs) hold significant promise in bone tissue engineering due to their self-renewal capacity and easy accessibility. However, their limited osteogenic potential remains a critical challenge for clinical application in bone repair. Emerging evidence suggests that circular RNAs (circRNAs) play a key role in regulating stem cell fate and osteogenesis. Despite this, the specific mechanisms by which circRNAs influence ADSCs in the context of bone tissue engineering are largely unexplored. This study introduces a novel strategy utilizing circAars, a specific circRNA, to modify ADSCs, which are then incorporated into gelatin methacryloyl (GelMA) hydrogels for the repair of critical-sized maxillofacial bone defects. The findings reveal that circAars predominantly localizes in the cytoplasm of ADSCs, where it acts as a competitive sponge for miR-128-3p, enhancing the osteogenic differentiation and migration capabilities of ADSCs. Furthermore, circAars-engineered ADSCs facilitate macrophage polarization from the M1 to M2 phenotype and enhance endothelial cell (EC) angiogenic potential through a paracrine mechanism. Additionally, GelMA scaffolds loaded with circAars-engineered ADSCs accelerate the repair of critical-sized maxillofacial bone defects by synergistically promoting osteogenesis, macrophage M2 polarization, and angiogenesis. This approach offers a promising therapeutic strategy for the treatment of critical-sized maxillofacial defects.
Collapse
Affiliation(s)
- Yi He
- Hospital of StomatologyGuanghua School of StomatologyInstitute of Stomatological ResearchSun Yat‐sen UniversityGuangZhou510080P. R. China
- Guangdong Provincial Key Laboratory of StomatologyGuangzhou510055P. R. China
| | - Yunyang Lu
- Hospital of StomatologyGuanghua School of StomatologyInstitute of Stomatological ResearchSun Yat‐sen UniversityGuangZhou510080P. R. China
- Guangdong Provincial Key Laboratory of StomatologyGuangzhou510055P. R. China
| | - Runze Li
- Hospital of StomatologyGuanghua School of StomatologyInstitute of Stomatological ResearchSun Yat‐sen UniversityGuangZhou510080P. R. China
- Guangdong Provincial Key Laboratory of StomatologyGuangzhou510055P. R. China
| | - Yuquan Tang
- Hospital of StomatologyGuanghua School of StomatologyInstitute of Stomatological ResearchSun Yat‐sen UniversityGuangZhou510080P. R. China
- Guangdong Provincial Key Laboratory of StomatologyGuangzhou510055P. R. China
- Guangzhou Liwan District Stomatological HospitalGuangzhou510080P. R. China
| | - Weidong Du
- Hospital of StomatologyGuanghua School of StomatologyInstitute of Stomatological ResearchSun Yat‐sen UniversityGuangZhou510080P. R. China
- Guangdong Provincial Key Laboratory of StomatologyGuangzhou510055P. R. China
| | - Lejia Zhang
- Hospital of StomatologyGuanghua School of StomatologyInstitute of Stomatological ResearchSun Yat‐sen UniversityGuangZhou510080P. R. China
- Guangdong Provincial Key Laboratory of StomatologyGuangzhou510055P. R. China
| | - Jie Wu
- Hospital of StomatologyGuanghua School of StomatologyInstitute of Stomatological ResearchSun Yat‐sen UniversityGuangZhou510080P. R. China
- Guangdong Provincial Key Laboratory of StomatologyGuangzhou510055P. R. China
| | - Kechen Li
- Hospital of StomatologyGuanghua School of StomatologyInstitute of Stomatological ResearchSun Yat‐sen UniversityGuangZhou510080P. R. China
- Guangdong Provincial Key Laboratory of StomatologyGuangzhou510055P. R. China
| | - Weijie Zhuang
- Hospital of StomatologyGuanghua School of StomatologyInstitute of Stomatological ResearchSun Yat‐sen UniversityGuangZhou510080P. R. China
- Guangdong Provincial Key Laboratory of StomatologyGuangzhou510055P. R. China
| | - Shiyu Lv
- Hospital of StomatologyGuanghua School of StomatologyInstitute of Stomatological ResearchSun Yat‐sen UniversityGuangZhou510080P. R. China
- Guangdong Provincial Key Laboratory of StomatologyGuangzhou510055P. R. China
| | - Yaoling Han
- Hospital of StomatologyGuanghua School of StomatologyInstitute of Stomatological ResearchSun Yat‐sen UniversityGuangZhou510080P. R. China
- Guangdong Provincial Key Laboratory of StomatologyGuangzhou510055P. R. China
| | - Bailong Tao
- Hospital of StomatologyGuanghua School of StomatologyInstitute of Stomatological ResearchSun Yat‐sen UniversityGuangZhou510080P. R. China
- Guangdong Provincial Key Laboratory of StomatologyGuangzhou510055P. R. China
- Laboratory Research CenterThe First Affiliated Hospital of Chongqing Medical UniversityChongqing400016P. R. China
| | - Feilong Deng
- Hospital of StomatologyGuanghua School of StomatologyInstitute of Stomatological ResearchSun Yat‐sen UniversityGuangZhou510080P. R. China
- Guangdong Provincial Key Laboratory of StomatologyGuangzhou510055P. R. China
| | - Wei Zhao
- Hospital of StomatologyGuanghua School of StomatologyInstitute of Stomatological ResearchSun Yat‐sen UniversityGuangZhou510080P. R. China
- Guangdong Provincial Key Laboratory of StomatologyGuangzhou510055P. R. China
| | - Dongsheng Yu
- Hospital of StomatologyGuanghua School of StomatologyInstitute of Stomatological ResearchSun Yat‐sen UniversityGuangZhou510080P. R. China
- Guangdong Provincial Key Laboratory of StomatologyGuangzhou510055P. R. China
| |
Collapse
|
14
|
Feng J, Wang F, Shao Y, Jin A, Lei L. Engineered protein-based materials for tissue repair: A review. Int J Biol Macromol 2025; 303:140674. [PMID: 39909268 DOI: 10.1016/j.ijbiomac.2025.140674] [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: 12/09/2024] [Revised: 01/19/2025] [Accepted: 02/02/2025] [Indexed: 02/07/2025]
Abstract
The human body may suffer multiple injuries and losses due to various external factors, such as tumors, diseases, traffic accidents, and war conflicts. Under such circumstances, engineered protein-based materials, as an innovative adjunctive material, can not only effectively promote the natural repair process of tissues, but also greatly circumvent the negative effects and complications that may be associated with conventional surgery. In this review, we first trace the definition and development of engineered protein-based materials and explain in detail their mechanism of action in promoting tissue repair. Subsequently, the advantages and disadvantages of various engineered protein-based materials in tissue repair are analyzed by comparison. In addition, the present review reveals in depth how material properties can be optimized by scientific means to meet different tissue repair needs. In addition, we present in detail specific application cases of engineered protein-based materials in the field of tissue repair. Finally, we summarize current challenges in engineered protein-based materials and provide an outlook for the future. This review not only provides theoretical support for the further exploration and development of engineered protein-based materials in the field of tissue repair, but also provides valuable references and inspiration for research in related fields.
Collapse
Affiliation(s)
- Jiayin Feng
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Institute of Translational Medicine, Zhejiang Shuren University, Hangzhou 310015, China
| | - Fangyan Wang
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Institute of Translational Medicine, Zhejiang Shuren University, Hangzhou 310015, China
| | - Yunyuan Shao
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Institute of Translational Medicine, Zhejiang Shuren University, Hangzhou 310015, China
| | - Anqi Jin
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Institute of Translational Medicine, Zhejiang Shuren University, Hangzhou 310015, China
| | - Lanjie Lei
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Institute of Translational Medicine, Zhejiang Shuren University, Hangzhou 310015, China.
| |
Collapse
|
15
|
Li W, Zhang H, Chen L, Huang C, Jiang Z, Zhou H, Zhu X, Liu X, Zheng Z, Yu Q, He Y, Gao Y, Ma J, Yang L. Cell membrane-derived nanovesicles as extracellular vesicle-mimetics in wound healing. Mater Today Bio 2025; 31:101595. [PMID: 40104636 PMCID: PMC11914519 DOI: 10.1016/j.mtbio.2025.101595] [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: 12/09/2024] [Revised: 01/28/2025] [Accepted: 02/17/2025] [Indexed: 03/20/2025] Open
Abstract
Cell membrane-derived nanovesicles (NVs) have emerged as promising alternatives to extracellular vesicles (EVs) for wound healing applications, addressing the limitations of traditional EVs, which include insufficient targeting capability, low production yield, and limited drug-loading capacity. Through mechanical cell extrusion methods, NVs exhibit superior characteristics, demonstrating enhanced yield, stability, and purity compared to natural EVs. These NVs can be derived from various membrane sources, including single cell types (stem cells, blood cells, immune cells, and bacterial membranes), hybrid cell membranes and cell membranes mixed with liposomes, with each offering unique therapeutic properties. The integration of genetic engineering and surface modifications has further enhanced NV functionality, enabling precise targeting and improved drug delivery capabilities. Recent advances in NV-based therapies have demonstrated their potential across multiple biomedical applications. Although challenges persist in terms of standardization, storage stability, and clinical translation, the combination of natural cell-derived functions with artificial modification potential positions NVs as a promising platform for next-generation therapeutic delivery systems, thereby offering new possibilities in wound healing applications. Finally, we explore the challenges and future prospects of translating NV-based therapeutics into clinical practice, providing insights into the future development of this innovative approach in wound healing and tissue repair.
Collapse
Affiliation(s)
- Wenwen Li
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Huihui Zhang
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Lianglong Chen
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Chaoyang Huang
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Ziwei Jiang
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Hai Zhou
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Xinxi Zhu
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Xiaoyang Liu
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Zesen Zheng
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Qiuyi Yu
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Yufang He
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Yanbin Gao
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Jun Ma
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Lei Yang
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| |
Collapse
|
16
|
Zhou J, Sun Z, Wang X, Wang S, Jiang W, Tang D, Xia T, Xiao F. Low-temperature cold plasma promotes wound healing by inhibiting skin inflammation and improving skin microbiome. Front Bioeng Biotechnol 2025; 13:1511259. [PMID: 40051835 PMCID: PMC11882593 DOI: 10.3389/fbioe.2025.1511259] [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: 10/14/2024] [Accepted: 02/03/2025] [Indexed: 03/09/2025] Open
Abstract
Wound healing includes four consecutive and overlapping stages of hemostasis, inflammation, proliferation, and remodeling. Factors such as aging, infection, and chronic diseases can lead to chronic wounds and delayed healing. Low-temperature cold plasma (LTCP) is an emerging physical therapy for wound healing, characterized by its safety, environmental friendliness, and ease of operation. This study utilized a self-developed LTCP device to investigate its biological effects and mechanisms on wound healing in adult and elderly mice. Histopathological studies found that LTCP significantly accelerated the healing rate of skin wounds in mice, with particularly pronounced effects in elderly mice. LTCP can markedly inhibit the expression of pro-inflammatory cytokines (TNF-α, IL-6, IL-1β) and senescence-associated secretory phenotype factors (MMP-3, MMP-9), while significantly increasing the expression of tissue repair-related factors, such as VEGF, bFGF, TGF-β, COL-I, and α-SMA. It also regulated the expression of genes related to cell proliferation and migration (Aqp5, Spint1), inflammation response (Nlrp3, Icam1), and angiogenesis (Ptx3, Thbs1), promoting cell proliferation and inhibit apoptosis. Furthermore, LTCP treatment reduced the relative abundance of harmful bacteria such as Delftia, Stenotrophomonas, Enterococcus, and Enterobacter in skin wounds, while increasing the relative abundance of beneficial bacteria such as Muribaculaceae, Acinetobacter, Lachnospiraceae NK4A136_group, and un_f__Lachnospiraceae, thereby improving the microbial community structure of skin wounds. These research findings are of significant implications for understanding the mechanism of skin wound healing, as well as for the treatment and clinical applications of skin wounds, especially aging skin.
Collapse
Affiliation(s)
- Jie Zhou
- School of Bioengineering, Qilu University of Technology (Shandong Academy of Science), Jinan, Shandong, China
- State Key Laboratory of Biobased Material and Green Papermaking, Qilu University of Technology (Shandong Academy of Science), Jinan, Shandong, China
| | - Zengkun Sun
- School of Bioengineering, Qilu University of Technology (Shandong Academy of Science), Jinan, Shandong, China
- State Key Laboratory of Biobased Material and Green Papermaking, Qilu University of Technology (Shandong Academy of Science), Jinan, Shandong, China
| | - Xiaoru Wang
- School of Bioengineering, Qilu University of Technology (Shandong Academy of Science), Jinan, Shandong, China
- State Key Laboratory of Biobased Material and Green Papermaking, Qilu University of Technology (Shandong Academy of Science), Jinan, Shandong, China
| | - Shouguo Wang
- Academy of Advanced Interdisciplinary Studies, Qilu University of Technology (Shandong Academy of Science), Jinan, Shandong, China
| | - Wen Jiang
- Beijing Zhongsu Titanium Alloy Vacuum Plasma Technology Research Institute, Beijing, China
| | - Dongqi Tang
- Center for Gene and Immunotherapy, Multidisciplinary Innovation Center for Nephrology, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Tao Xia
- School of Bioengineering, Qilu University of Technology (Shandong Academy of Science), Jinan, Shandong, China
- State Key Laboratory of Biobased Material and Green Papermaking, Qilu University of Technology (Shandong Academy of Science), Jinan, Shandong, China
| | - Fang Xiao
- Department of Gerontology, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| |
Collapse
|
17
|
Yang J, Yuan J, Wen YQ, Wu L, Liao JJ, Qi HB. Bone marrow mesenchymal stem cells promote uterine healing by activating the PI3K/AKT pathway and modulating inflammation in rat models. World J Stem Cells 2025; 17:98349. [PMID: 39866893 PMCID: PMC11752458 DOI: 10.4252/wjsc.v17.i1.98349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 10/02/2024] [Accepted: 12/10/2024] [Indexed: 01/20/2025] Open
Abstract
BACKGROUND Uterine injury can cause uterine scarring, leading to a series of complications that threaten women's health. Uterine healing is a complex process, and there are currently no effective treatments. Although our previous studies have shown that bone marrow mesenchymal stem cells (BMSCs) promote uterine damage repair, the underlying mechanisms remain unclear. However, exploring the specific regulatory roles of BMSCs in uterine injury treatment is crucial for further understanding their functions and enhancing therapeutic efficacy. AIM To investigate the underlying mechanism by which BMSCs promote the process of uterine healing. METHODS In in vivo experiments, we established a model of full-thickness uterine injury and injected BMSCs into the uterine wound. Transcriptome sequencing was performed to determine the enrichment of differentially expressed genes at the wound site. In in vitro experiments, we isolated rat uterine smooth muscle cells (USMCs) and cocultured them with BMSCs to observe the interaction between BMSCs and USMCs in the microenvironment. RESULTS We found that the differentially expressed genes were mainly related to cell growth, tissue repair, and angiogenesis, while the phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT) pathway was highly enriched. Quantitative reverse-transcription polymerase chain reaction was used to validate differentially expressed genes, and the results demonstrated that BMSCs can upregulate genes related to regeneration and downregulate genes related to inflammation. Coculturing BMSCs promoted the migration and proliferation of USMCs, and the USMC microenvironment promoted the myogenic differentiation of BMSCs. Finally, we validated the PI3K/AKT pathway in tissues and cells and showed that BMSCs activate the PI3K/AKT pathway to promote the regeneration of uterine smooth muscle both in vivo and in vitro. CONCLUSION BMSCs upregulated uterine wound regeneration and anti-inflammatory factors and enhanced uterine smooth muscle proliferation through the PI3K/AKT pathway both in vivo and in vitro.
Collapse
Affiliation(s)
- Jing Yang
- Women and Children's Hospital of Chongqing Medical University, Chongqing 401147, China
- Chongqing Key Laboratory of Maternal and Fetal Medicine, Chongqing Medical University, Chongqing 400016, China
- Obstetrics and Gynecology, Guizhou Provincial People's Hospital, Guiyang 557300, Guizhou Province, China
| | - Jun Yuan
- Women and Children's Hospital of Chongqing Medical University, Chongqing 401147, China
- Chongqing Key Laboratory of Maternal and Fetal Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Yan-Qing Wen
- Women and Children's Hospital of Chongqing Medical University, Chongqing 401147, China
- Chongqing Key Laboratory of Maternal and Fetal Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Li Wu
- Women and Children's Hospital of Chongqing Medical University, Chongqing 401147, China
| | - Jiu-Jiang Liao
- Women and Children's Hospital of Chongqing Medical University, Chongqing 401147, China
- Chongqing Key Laboratory of Maternal and Fetal Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Hong-Bo Qi
- Women and Children's Hospital of Chongqing Medical University, Chongqing 401147, China
- Chongqing Key Laboratory of Maternal and Fetal Medicine, Chongqing Medical University, Chongqing 400016, China
- Research Laboratory of Reproduction and Development of Chinese Ministry of Education, Chongqing Medical University, Chongqing 400016, China.
| |
Collapse
|
18
|
Wolint P, Hofmann S, von Atzigen J, Böni R, Miescher I, Giovanoli P, Calcagni M, Emmert MY, Buschmann J. Standardization to Characterize the Complexity of Vessel Network Using the Aortic Ring Model. Int J Mol Sci 2024; 26:291. [PMID: 39796147 PMCID: PMC11719671 DOI: 10.3390/ijms26010291] [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: 11/13/2024] [Revised: 12/05/2024] [Accepted: 12/30/2024] [Indexed: 01/13/2025] Open
Abstract
Regeneration after ischemia requires to be promoted by (re)perfusion of the affected tissue, and, to date, there is no therapy that covers all needs. In treatment with mesenchymal stem cells (MSC), the secretome acts via paracrine mechanisms and has a positive influence on vascular regeneration via proangiogenic factors. A lack of standardization and the high complexity of vascular structures make it difficult to compare angiogenic readouts from different studies. This emphasizes the need for improved approaches and the introduction of an index in the preclinical setting. A characterization of human MSC secretomes obtained from one of the three formats-single cells, small, and large spheroids-was performed using the chicken aortic ring assay in combination with a modified angiogenic activity index (AAI) and an angiogenic profile. While the secretome of the small spheroid group showed an inhibitory effect on angiogenesis, the large spheroid group impressed with a fully pro-angiogenic response, and a higher AAI compared to the single cell group, underlying the suitability of these three-stem cell-derived secretomes with their distinct angiogenic properties to validate the AAI and the novel angiogenic profile established here.
Collapse
Affiliation(s)
- Petra Wolint
- Division of Surgical Research, University Hospital of Zurich, 8091 Zurich, Switzerland
- Department of Plastic Surgery and Hand Surgery, University Hospital Zurich, 8091 Zurich, Switzerland; (S.H.); (J.v.A.); (I.M.); (P.G.); (M.C.)
| | - Silvan Hofmann
- Department of Plastic Surgery and Hand Surgery, University Hospital Zurich, 8091 Zurich, Switzerland; (S.H.); (J.v.A.); (I.M.); (P.G.); (M.C.)
| | - Julia von Atzigen
- Department of Plastic Surgery and Hand Surgery, University Hospital Zurich, 8091 Zurich, Switzerland; (S.H.); (J.v.A.); (I.M.); (P.G.); (M.C.)
| | - Roland Böni
- White House Center for Liposuction, 8044 Zurich, Switzerland;
| | - Iris Miescher
- Department of Plastic Surgery and Hand Surgery, University Hospital Zurich, 8091 Zurich, Switzerland; (S.H.); (J.v.A.); (I.M.); (P.G.); (M.C.)
| | - Pietro Giovanoli
- Department of Plastic Surgery and Hand Surgery, University Hospital Zurich, 8091 Zurich, Switzerland; (S.H.); (J.v.A.); (I.M.); (P.G.); (M.C.)
| | - Maurizio Calcagni
- Department of Plastic Surgery and Hand Surgery, University Hospital Zurich, 8091 Zurich, Switzerland; (S.H.); (J.v.A.); (I.M.); (P.G.); (M.C.)
| | - Maximilian Y. Emmert
- Institute for Regenerative Medicine (IREM), University of Zurich, 8952 Zurich, Switzerland;
- Deutsches Herzzentrum der Charité (DHZC), Department of Cardiothoracic and Vascular Surgery, 13353 Berlin, Germany
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 13353 Berlin, Germany
- BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health at Charité-Universitätsmedizin Berlin, 13353 Berlin, Germany
| | - Johanna Buschmann
- Division of Surgical Research, University Hospital of Zurich, 8091 Zurich, Switzerland
- Department of Plastic Surgery and Hand Surgery, University Hospital Zurich, 8091 Zurich, Switzerland; (S.H.); (J.v.A.); (I.M.); (P.G.); (M.C.)
| |
Collapse
|
19
|
Nifontova G, Safaryan S, Khristidis Y, Smirnova O, Vosough M, Shpichka A, Timashev P. Advancing wound healing by hydrogel-based dressings loaded with cell-conditioned medium: a systematic review. Stem Cell Res Ther 2024; 15:371. [PMID: 39420416 PMCID: PMC11488269 DOI: 10.1186/s13287-024-03976-x] [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: 10/04/2024] [Indexed: 10/19/2024] Open
Abstract
BACKGROUND Wound healing represents a complex biological process, critically important in clinical practice due to its direct implication in a patient's recovery and quality of life. Conservative wound management frequently falls short in providing an ideal environment for the optimal tissue regeneration, often resulting in extended healing periods and elevated risk of infection and other complications. The emerging biomaterials, particularly hydrogels, have shown substantial promise in addressing these challenges by offering properties such as biocompatibility, biodegradability, and the ability to cure wound environment. Recent advancements have highlighted the therapeutic potential of integrating cell-derived conditioned medium (CM) into hydrogel matrices. Cell-derived CM represents a rich array of bioactive molecules, demonstrating significant efficacy in modulating cellular activities crucial for wound healing, including cellular proliferation, migration, and angiogenesis. METHODS The methodology of this review adheres to the standards set by the Preferred Reporting Items for Systematic Review and Meta-Analysis (PRISMA) guidelines. The review includes a selection of studies published within the last five years, focusing on in vivo experiments involving various types of skin injuries treated with topically applied hydrogels loaded with CM (H-CM). The search strategy refers to the PICO framework and includes the assessment of study quality by CAMARADES tool. RESULTS The systematic review represents a detailed evaluation of H-CM dressings wound healing efficiency based on the experimental results of cell-based assays and animal wound models. The study targets to reveal wound healing capacity of H-CM dressings, and provides a comparative data analysis, limitations of methods and discussions of H-CM role in advancing the wound healing therapy. CONCLUSIONS The data presented demonstrate that H-CM is a promising material for advanced wound healing and regenerative medicine. These dressings possess proved in vitro/in vivo efficacy that highlights their strong clinical potential and paves the way to further investigations of H-CM formulations within clinical trials.
Collapse
Affiliation(s)
- Galina Nifontova
- Institute for Regenerative Medicine, Sechenov University, 8-2 Trubetskaya St, Moscow, 119991, Russia
| | - Sofia Safaryan
- Institute for Regenerative Medicine, Sechenov University, 8-2 Trubetskaya St, Moscow, 119991, Russia
| | - Yana Khristidis
- Institute for Regenerative Medicine, Sechenov University, 8-2 Trubetskaya St, Moscow, 119991, Russia
| | - Olga Smirnova
- Institute for Regenerative Medicine, Sechenov University, 8-2 Trubetskaya St, Moscow, 119991, Russia
| | - Massoud Vosough
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, Academic Center for Education, Culture and Research, Tehran, 1665666311, Iran
| | - Anastasia Shpichka
- Institute for Regenerative Medicine, Sechenov University, 8-2 Trubetskaya St, Moscow, 119991, Russia.
| | - Peter Timashev
- Institute for Regenerative Medicine, Sechenov University, 8-2 Trubetskaya St, Moscow, 119991, Russia
- World-Class Research Center "Digital Biodesign and Personalized Healthcare", Sechenov University, 8-2 Trubetskaya St, Moscow, 119991, Russia
| |
Collapse
|
20
|
Tanadchangsaeng N, Pasanaphong K, Tawonsawatruk T, Rattanapinyopituk K, Tangketsarawan B, Rawiwet V, Kongchanagul A, Srikaew N, Yoyruerop T, Panupinthu N, Sangpayap R, Panaksri A, Boonyagul S, Hemstapat R. 3D bioprinting of fish skin-based gelatin methacryloyl (GelMA) bio-ink for use as a potential skin substitute. Sci Rep 2024; 14:23240. [PMID: 39369014 PMCID: PMC11455937 DOI: 10.1038/s41598-024-73774-1] [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: 07/19/2024] [Accepted: 09/20/2024] [Indexed: 10/07/2024] Open
Abstract
Gelatin methacryloyl (GelMA), typically derived from mammalian sources, has recently emerged as an ideal bio-ink for three-dimensional (3D) bioprinting. Herein, we developed a fish skin-based GelMA bio-ink for the fabrication of a 3D GelMA skin substitute with a 3D bioprinter. Several concentrations of methacrylic acid anhydride were used to fabricate GelMA, in which their physical-mechanical properties were assessed. This fish skin-based GelMA bio-ink was loaded with human adipose tissue-derived mesenchymal stromal cells (ASCs) and human platelet lysate (HPL) and then printed to obtain 3D ASCs + HPL-loaded GelMA scaffolds. Cell viability test and a preliminary investigation of its effectiveness in promoting wound closure were evaluated in a critical-sized full thickness skin defect in a rat model. The cell viability results showed that the number of ASCs increased significantly within the 3D GelMA hydrogel scaffold, indicating its biocompatibility property. In vivo results demonstrated that ASCs + HPL-loaded GelMA scaffolds could delay wound contraction, markedly enhanced collagen deposition, and promoted the formation of new blood vessels, especially at the wound edge, compared to the untreated group. Therefore, this newly fish skin-based GelMA bio-ink developed in this study has the potential to be utilized for the printing of 3D GelMA skin substitutes.
Collapse
Affiliation(s)
| | | | - Tulyapruek Tawonsawatruk
- Department of Orthopaedics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Kasem Rattanapinyopituk
- Department of Pathology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand
| | | | - Visut Rawiwet
- Central Animal Facility, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Alita Kongchanagul
- Center for Vaccine Development, Institute of Molecular Biosciences, Mahidol University, Bangkok, Thailand
| | - Narongrit Srikaew
- Research Center, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Thanaporn Yoyruerop
- Mahidol University-Frontier Research Facility (MU-FRF), Mahidol University, Nakhon Pathom, Thailand
| | - Nattapon Panupinthu
- Department of Physiology, Faculty of Science, Mahidol University, Bangkok, Thailand
- Center of Calcium and Bone Research (COCAB), Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Ratirat Sangpayap
- Department of Pharmacology, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Anuchan Panaksri
- College of Biomedical Engineering, Rangsit University, Pathum Thani, Thailand
| | - Sani Boonyagul
- College of Biomedical Engineering, Rangsit University, Pathum Thani, Thailand
| | - Ruedee Hemstapat
- Department of Pharmacology, Faculty of Science, Mahidol University, Bangkok, Thailand.
| |
Collapse
|
21
|
Asadi K, Azarpira N, Heidari R, Hamidi M, Yousefzadeh-Chabok S, Nemati MM, Ommati MM, Amini A, Gholami A. Trinitroglycerin-loaded chitosan nanogels accelerate angiogenesis in wound healing process. Int J Biol Macromol 2024; 278:134937. [PMID: 39179074 DOI: 10.1016/j.ijbiomac.2024.134937] [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: 06/14/2024] [Revised: 08/14/2024] [Accepted: 08/20/2024] [Indexed: 08/26/2024]
Abstract
Trinitroglycerin (TNG) with remarkable angiogenic, antibacterial, and antioxidative activity is a promising candidate to govern wound healing capacity. However, its clinical administration is limited due to associated complications and NO short half-life. In the current study, TNG-loaded chitosan nanogels (TNG-Ngs) were examined in-vitro and in-vivo to gain insight into their clinical application. We prepared TNG-Ngs and characterized their physiochemical properties. The potential of TNG-Ngs was assessed using biocompatibility, scratch assay, and a full-thickness skin wounds model, followed by histopathological and immunohistochemistry examinations. TNG-Ngs particle size 96 ± 18 and definite size distribution histogram. The loading capacity (LC) and encapsulation efficiency (EE) of prepared TNG-Ngs were 70.2 % and 2.1 %, respectively. The TNG-Ngs samples showed enhanced migration of HUVECs with no apparent cytotoxicity. The topical use of TNG-Ngs200 on the wounds revealed a complete wound closure ratio, skin component formation, less scar width, remarkable granulation tissue, promoted collagen deposition, and enhanced the relative mean density of α-SMA and CD31. TNG-Ngs accelerated wound healing by promoting collagen deposition and angiogenic activity, as well as reducing inflammation. The findings indicated that TNG-Ngs is expected to be well vascularized in the wound area and to be more effective in topical therapy.
Collapse
Affiliation(s)
- Khatereh Asadi
- Biotechnology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran; Department of Medical Nanotechnology, School of Advanced Medical Science and Technology, Shiraz University of Medical Sciences, Shiraz, Iran; Guilan Road Trauma Research Center, Guilan University of Medical Sciences, Rasht, Iran
| | - Negar Azarpira
- Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Reza Heidari
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mehrdad Hamidi
- Department of Pharmaceutics, School of Pharmacy, Zanjan University of Medical Sciences, 45139-56184 Zanjan, Iran
| | | | - Mohammad Mehdi Nemati
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Mehdi Ommati
- Henan Key Laboratory of Environmental and Animal Product Safety, College of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471000, Henan, China
| | - Abbas Amini
- Abdullah Al Salem University (AASU), College of Engineering and Energy, Khaldiya, Kuwait; Centre for Infrastructure Engineering, Western Sydney University, Penrith, NSW, Australia
| | - Ahmad Gholami
- Biotechnology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran; Department of Medical Nanotechnology, School of Advanced Medical Science and Technology, Shiraz University of Medical Sciences, Shiraz, Iran; Department of Pharmaceutical Biotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
22
|
Ai C, Bai J, Ye Q, Niu S, Li Y, Li P, Wu H, Wu J, Wang X. Accelerating healing at high altitudes: Oxygen and bFGF delivery through nanoparticle-loaded gel dressings. Biomed Pharmacother 2024; 179:117247. [PMID: 39236477 DOI: 10.1016/j.biopha.2024.117247] [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: 05/06/2024] [Revised: 07/12/2024] [Accepted: 07/30/2024] [Indexed: 09/07/2024] Open
Abstract
High altitude environment is mainly characterized by low oxygen. Due to persistent hypoxia, nonhealing wounds are common in high-altitude areas. Moreover, Basic fibroblast growth factor (bFGF) is a versatile biologically active substance that has crucial impact on wound healing. Given the limited availability of atmospheric oxygen and reduced blood oxygen saturation in high-altitude area, and the challenge that arises from direct oxygen and bFGF delivery to wounds through the traumatized vascular structure, it necessitates an innovative solution for local and permeable delivery of oxygen and bFGF. In this study, we present a strategy that involves revamping traditional gel-based wound dressings through the incorporation of nanoparticles encapsulating oxygen and bFGF, engineered to facilitate the localized delivery of dissolved oxygen and bFGF to wound surfaces. The prospective evaluation of this delivery technique's therapeutic impacts on epithelial, endothelial and fibroblasts cells can be materialized. Further experiment corroborated these effects on a high-altitude wounds' murine model. Given its biocompatibility, efficacy, and utility, we posit that NOB-Gel exhibits remarkable translational potential for managing and hastening the healing process of an array of clinical wounds, more so for wounds inflicted at high altitudes.
Collapse
Affiliation(s)
- Chongyi Ai
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China
| | - Jin Bai
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China; School and Hospital of Stomatology, Tianjin Medical University, Tianjin 300070, China
| | - Qingsong Ye
- State Key Laboratory of Pharmaceutical Biotechnology, Chemistry and Biomedicine Innovation Center, Medical School of Nanjing University, Nanjing 210093, China
| | - Siyu Niu
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China
| | - Yunzhe Li
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China; School and Hospital of Stomatology, Tianjin Medical University, Tianjin 300070, China
| | - Pan Li
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China; School and Hospital of Stomatology, Tianjin Medical University, Tianjin 300070, China
| | - Hao Wu
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China
| | - Jinhui Wu
- State Key Laboratory of Pharmaceutical Biotechnology, Chemistry and Biomedicine Innovation Center, Medical School of Nanjing University, Nanjing 210093, China.
| | - Xinxing Wang
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China.
| |
Collapse
|
23
|
Zhu Y, Liu X, Chen X, Liao Y. Adipose-derived stem cells apoptosis rejuvenate radiation-impaired skin in mice via remodeling and rearranging dermal collagens matrix. Stem Cell Res Ther 2024; 15:324. [PMID: 39334464 PMCID: PMC11438223 DOI: 10.1186/s13287-024-03904-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 08/26/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND Chronic radiation dermatitis (CRD) is a late consequence of radiation with high incidence in patients receiving radiotherapy. Conventional therapies often yield unsatisfactory results. Therefore, this study aimed to explore the therapeutic potential and mechanism of adipose-derived stem cells (ADSCs) for CRD, paving the way for novel regenerative therapies in clinical practice. METHODS Clinical CRD skin biopsies were analyzed to character the pathological changes of CRD skin and guided the animal modeling scheme. Subsequently, an in vivo analysisusing mouse CRD models was conducted to explore their effects of ADSCs on CRD, monitoring therapeutic impact for up to 8 weeks. Transcriptome sequencing and histologic sections analysis were performed to explore the potential therapeutic mechanism of ADSCs. Following observing extensive apoptosis of transplanted ADSCs, the therapeutic effect of ADSCs were compared with those of apoptosis-inhibited ADSCs. Multiphoton imaging and analysis of collagen morphologic features were employed to explain how translated ADSCs promote collagen remodeling at the microscopic level based on the contrast of morphology of collagen fibers. RESULTS Following injection into CRD-afflicted skin, ADSCs therapy effectively mitigated symptoms of CRD, including acanthosis of the epidermis, fibrosis, and irregular collagen deposition, consistent with the possible therapeutic mechanism suggested by transcriptome sequencing. Notably, in vivo tracking revealed a significant reduction in ADSCs number due to extensive apoptosis. Inhibiting apoptosis in ADSCs partially tempered their therapeutic effects. Mechanically, analysis of collagen morphologic features indicated that translated ADSCs might promote dermal extracellular matrix remodeling through enlarging, lengthening, crimping, and evening collagen, counteracting the atrophy and rupture caused by irradiation. CONCLUSIONS This study demonstrated that ADSCs underwent substantial apoptosis upon local skin transplantation, and paradoxically, this apoptosis is essential for their efficacy in promoting the regeneration of late radiation-impaired skin. Mechanically, transplanted ADSCs could promote the remodeling and rearrangement of radiation-damaged dermal collagen matrix.
Collapse
Affiliation(s)
- Yufan Zhu
- Department of Plastic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, 510515, Guangdong, People's Republic of China
| | - Xu Liu
- Department of Plastic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, 510515, Guangdong, People's Republic of China
| | - Xihang Chen
- Department of Plastic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, 510515, Guangdong, People's Republic of China.
| | - Yunjun Liao
- Department of Plastic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, 510515, Guangdong, People's Republic of China.
| |
Collapse
|
24
|
Shu F, Huang H, Xiao S, Xia Z, Zheng Y. Netrin-1 co-cross-linked hydrogel accelerates diabetic wound healing in situ by modulating macrophage heterogeneity and promoting angiogenesis. Bioact Mater 2024; 39:302-316. [PMID: 38827174 PMCID: PMC11143790 DOI: 10.1016/j.bioactmat.2024.04.019] [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: 01/20/2024] [Revised: 04/16/2024] [Accepted: 04/18/2024] [Indexed: 06/04/2024] Open
Abstract
Diabetic wounds, characterized by prolonged inflammation and impaired vascularization, are a serious complication of diabetes. This study aimed to design a gelatin methacrylate (GelMA) hydrogel for the sustained release of netrin-1 and evaluate its potential as a scaffold to promote diabetic wound healing. The results showed that netrin-1 was highly expressed during the inflammation and proliferation phases of normal wounds, whereas it synchronously exhibited aberrantly low expression in diabetic wounds. Neutralization of netrin-1 inhibited normal wound healing, and the topical application of netrin-1 accelerated diabetic wound healing. Mechanistic studies demonstrated that netrin-1 regulated macrophage heterogeneity via the A2bR/STAT/PPARγ signaling pathway and promoted the function of endothelial cells, thus accelerating diabetic wound healing. These data suggest that netrin-1 is a potential therapeutic target for diabetic wounds.
Collapse
Affiliation(s)
- Futing Shu
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, 200433, People's Republic of China
| | - Hongchao Huang
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, 200433, People's Republic of China
| | - Shichu Xiao
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, 200433, People's Republic of China
| | - Zhaofan Xia
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, 200433, 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, Shanghai, 200433, People's Republic of China
| | - Yongjun Zheng
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, 200433, People's Republic of China
| |
Collapse
|
25
|
Jin T, Fu Z, Zhou L, Chen L, Wang J, Wang L, Yan S, Li T, Jin P. GelMA loaded with platelet lysate promotes skin regeneration and angiogenesis in pressure ulcers by activating STAT3. Sci Rep 2024; 14:18345. [PMID: 39112598 PMCID: PMC11306777 DOI: 10.1038/s41598-024-67304-2] [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: 01/02/2024] [Accepted: 07/10/2024] [Indexed: 08/10/2024] Open
Abstract
Pressure ulcers (PU) are caused by persistent long-term pressure, which compromises the integrity of the epidermis, dermis, and subcutaneous adipose tissue layer by layer, making it difficult to heal. Platelet products such as platelet lysate (PL) can promote tissue regeneration by secreting numerous growth factors based on clinical studies on skin wound healing. However, the components of PL are difficult to retain in wounds. Gelatin methacrylate (GelMA) is a photopolymerizable hydrogel that has lately emerged as a promising material for tissue engineering and regenerative medicine. The PL liquid was extracted, flow cytometrically detected for CD41a markers, and evenly dispersed in the GelMA hydrogel to produce a surplus growth factor hydrogel system (PL@GM). The microstructure of the hydrogel system was observed under a scanning electron microscope, and its sustained release efficiency and biological safety were tested in vitro. Cell viability and migration of human dermal fibroblasts, and tube formation assays of human umbilical vein endothelial cells were applied to evaluate the ability of PL to promote wound healing and regeneration in vitro. Real-time polymerase chain reaction (PCR) and western blot analyses were performed to elucidate the skin regeneration mechanism of PL. We verified PL's therapeutic effectiveness and histological analysis on the PU model. PL promoted cell viability, migration, wound healing and angiogenesis in vitro. Real-time PCR and western blot indicated PL suppressed inflammation and promoted collagen I synthesis by activating STAT3. PL@GM hydrogel system demonstrated optimal biocompatibility and favorable effects on essential cells for wound healing. PL@GM also significantly stimulated PU healing, skin regeneration, and the formation of subcutaneous collagen and blood vessels. PL@GM could accelerate PU healing by promoting fibroblasts to migrate and secrete collagen and endothelial cells to vascularize. PL@GM promises to be an effective and convenient treatment modality for PU, like chronic wound treatment.
Collapse
Affiliation(s)
- Tingting Jin
- Center for Plastic and Reconstructive Surgery, Department of Plastic and Reconstructive Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, China
| | - Zexin Fu
- Center for Plastic and Reconstructive Surgery, Department of Plastic and Reconstructive Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, China
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Liuyi Zhou
- Center for Plastic and Reconstructive Surgery, Department of Plastic and Reconstructive Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, China
| | - Lulu Chen
- Center for Plastic and Reconstructive Surgery, Department of Plastic and Reconstructive Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, China
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Ji Wang
- Center for Plastic and Reconstructive Surgery, Department of Plastic and Reconstructive Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, China
| | - Lu Wang
- Center for Plastic and Reconstructive Surgery, Department of Plastic and Reconstructive Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, China
| | - Sheng Yan
- Center for Plastic and Reconstructive Surgery, Department of Plastic and Reconstructive Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, China
| | - Ting Li
- Center for Plastic and Reconstructive Surgery, Department of Plastic and Reconstructive Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, China.
| | - Peihong Jin
- Center for Plastic and Reconstructive Surgery, Department of Plastic and Reconstructive Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, China.
| |
Collapse
|
26
|
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
|
27
|
Santos N, Fuentes-Lemus E, Ahumada M. Use of photosensitive molecules in the crosslinking of biopolymers: applications and considerations in biomaterials development. J Mater Chem B 2024; 12:6550-6562. [PMID: 38913025 DOI: 10.1039/d4tb00299g] [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: 06/25/2024]
Abstract
The development of diverse types of biomaterials has significantly contributed to bringing new biomedical strategies to treat clinical conditions. Applications of these biomaterials can range from mechanical support and protection of injured tissues to joint replacement, tissue implants, and drug delivery systems. Among the strategies commonly used to prepare biomaterials, the use of electromagnetic radiation to initiate crosslinking stands out. The predominance of photo-induced polymerization methods relies on a fast, efficient, and straightforward process that can be easily adjusted to clinical needs. This strategy consists of irradiating the components that form the material with photons in the near ultraviolet-visible wavelength range (i.e., ∼310 to 750 nm) in the presence of a photoactive molecule. Upon photon absorption, photosensitive molecules can generate excited species that initiate photopolymerization through different reaction mechanisms. However, this process could promote undesired side reactions depending on the target zone or treatment type (e.g., oxidative stress and modification of biomolecules such as proteins and lipids). This review explores the basic concepts behind the photopolymerization process of ex situ and in situ biomaterials. Particular emphasis was put on the photosensitization initiated by the most employed photosensitizers and the photoreactions that they mediate in aqueous media. Finally, the undesired oxidation reactions at the bio-interface and potential solutions are presented.
Collapse
Affiliation(s)
- Nicolas Santos
- Institut Químic de Sarrià, Universitat Ramon Llull, Barcelona 08017, Spain
| | - Eduardo Fuentes-Lemus
- Department of Biomedical Sciences, Panum Institute, University of Copenhagen, Blegdamsvej 3, Copenhagen, 2200, Denmark.
| | - Manuel Ahumada
- Centro de Nanotecnología Aplicada, Facultad de Ciencias, Ingeniería y Tecnología, Universidad Mayor, Santiago 8580745, Chile.
- Escuela de Biotecnología, Facultad de Ciencias, Ingeniería y Tecnología, Universidad Mayor, Santiago 8580745, Chile
| |
Collapse
|
28
|
Zhou C, Cai Z, Guo J, Li C, Qin C, Yan J, Yang D. Injective hydrogel loaded with liposomes-encapsulated MY-1 promotes wound healing and increases tensile strength by accelerating fibroblast migration via the PI3K/AKT-Rac1 signaling pathway. J Nanobiotechnology 2024; 22:396. [PMID: 38965546 PMCID: PMC11225333 DOI: 10.1186/s12951-024-02666-3] [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] [Accepted: 06/25/2024] [Indexed: 07/06/2024] Open
Abstract
Failed skin wound healing, through delayed wound healing or wound dehiscence, is a global public health issue that imposes significant burdens on individuals and society. Although the application of growth factor is an effective method to improve the pace and quality of wound healing, the clinically approved factors are limited. Parathyroid hormone (PTH) demonstrates promising results in wound healing by promoting collagen deposition and cell migration, but its application is limited by potentially inhibitory effects when administered continuously and locally. Through partially replacing and repeating the amino acid domains of PTH(1-34), we previously designed a novel PTH analog, PTH(3-34)(29-34) or MY-1, and found that it avoided the inhibitory effects of PTH while retaining its positive functions. To evaluate its role in wound healing, MY-1 was encapsulated in liposomes and incorporated into the methacryloyl gelatin (GelMA) hydrogel, through which an injectable nanocomposite hydrogel (GelMA-MY@Lipo, or GML) was developed. In vitro studies revealed that the GML had similar properties in terms of the appearance, microstructure, functional groups, swelling, and degradation capacities as the GelMA hydrogel. In vitro drug release testing showed a relatively more sustainable release of MY-1, which was still detectable in vivo 9 days post-application. When the GML was topically applied to the wound areas of rat models, wound closure as well as tensile strength were improved. Further studies showed that the effects of GML on wound repair and tensile strength were closely related to the promotion of fibroblast migration to the wound area through the controlled release of MY-1. Mechanically, MY-1 enhanced fibroblast migration by activating PI3K/AKT signaling and its downstream molecule, Rac1, by which it increased fibroblast aggregation in the early stage and resulting in denser collagen deposition at a later time. Overall, these findings demonstrated that the nanocomposite hydrogel system promoted skin wound healing and increased tensile strength, thus offering new potential in the treatment of wound healing.
Collapse
Affiliation(s)
- Chunhao Zhou
- Department of Orthopedics - Spinal Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Avenue, Guangzhou, 510515, China
| | - Zhihai Cai
- Department of Orthopedics - Spinal Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Avenue, Guangzhou, 510515, China
| | - Jialiang Guo
- Department of Orthopedics - Spinal Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Avenue, Guangzhou, 510515, China
| | - Chengfu Li
- Department of Orthopedics - Spinal Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Avenue, Guangzhou, 510515, China
| | - Chenghe Qin
- Department of Orthopedics - Traumatology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Juanwen Yan
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| | - Dehong Yang
- Department of Orthopedics - Spinal Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Avenue, Guangzhou, 510515, China.
| |
Collapse
|
29
|
Lin Z, Lin D, Lin D. The Mechanisms of Adipose Stem Cell-Derived Exosomes Promote Wound Healing and Regeneration. Aesthetic Plast Surg 2024; 48:2730-2737. [PMID: 38438760 DOI: 10.1007/s00266-024-03871-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: 08/11/2023] [Accepted: 01/25/2024] [Indexed: 03/06/2024]
Abstract
Chronic wound healing is a class of diseases influenced by multiple complex factors, causing severe psychological and physiological impact on patients. It is an intractable clinical challenge and its possible mechanisms are not yet clear. It has been proven that adipose stem cell-derived exosomes (ADSC-Exos) can promote wound healing and inhibit scar formation by regulating inflammation, promoting cell proliferation, migration, and angiogenesis, regulating matrix remodeling, which provides a new approach for wound healing through biological treatment. This review focuses on the mechanism, treatment, and administration methods of ADSC-Exos in wound healing, providing a comprehensive understanding the mechanisms of ADSC-Exos on wound healing. LEVEL OF EVIDENCE I: This journal requires that authors assign a level of evidence to each article. 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)
- Zhengjie Lin
- Department of Metabolism and Endocrinology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Danyi Lin
- Department of Pathology, Guangdong Provincial People's Hospital, Southern Medical University, Guangzhou, China.
| | - Dane Lin
- Neonatal Intensive Care Unit, Department of Pediatrics, The First Affiliated Hospital of Shantou University Medical College, No. 57 Changping Road, Shantou, Guangdong, China.
| |
Collapse
|
30
|
Sabetkish S, Currie P, Meagher L. Recent trends in 3D bioprinting technology for skeletal muscle regeneration. Acta Biomater 2024; 181:46-66. [PMID: 38697381 DOI: 10.1016/j.actbio.2024.04.038] [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: 12/14/2023] [Revised: 04/24/2024] [Accepted: 04/25/2024] [Indexed: 05/05/2024]
Abstract
Skeletal muscle is a pro-regenerative tissue, that utilizes a tissue-resident stem cell system to effect repair upon injury. Despite the demonstrated efficiency of this system in restoring muscle mass after many acute injuries, in conditions of severe trauma such as those evident in volumetric muscle loss (VML) (>20 % by mass), this self-repair capability is unable to restore tissue architecture, requiring interventions which currently are largely surgical. As a possible alternative, the generation of artificial muscle using tissue engineering approaches may also be of importance in the treatment of VML and muscle diseases such as dystrophies. Three-dimensional (3D) bioprinting has been identified as a promising technique for regeneration of the complex architecture of skeletal muscle. This review discusses existing treatment strategies following muscle damage, recent progress in bioprinting techniques, the bioinks used for muscle regeneration, the immunogenicity of scaffold materials, and in vitro and in vivo maturation techniques for 3D bio-printed muscle constructs. The pros and cons of these bioink formulations are also highlighted. Finally, we present the current limitations and challenges in the field and critical factors to consider for bioprinting approaches to become more translationa and to produce clinically relevant engineered muscle. STATEMENT OF SIGNIFICANCE: This review discusses the physiopathology of muscle injuries and existing clinical treatment strategies for muscle damage, the types of bioprinting techniques that have been applied to bioprinting of muscle, and the bioinks commonly used for muscle regeneration. The pros and cons of these bioinks are highlighted. We present a discussion of existing gaps in the literature and critical factors to consider for the translation of bioprinting approaches and to produce clinically relevant engineered muscle. Finally, we provide insights into what we believe will be the next steps required before the realization of the application of tissue-engineered muscle in humans. We believe this manuscript is an insightful, timely, and instructive review that will guide future muscle bioprinting research from a fundamental construct creation approach, down a translational pathway to achieve the desired impact in the clinic.
Collapse
Affiliation(s)
- Shabnam Sabetkish
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia; ARC Training Centre for Cell and Tissue Engineering Technologies, Monash University, Clayton, VIC 3800, Australia
| | - Peter Currie
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia; ARC Training Centre for Cell and Tissue Engineering Technologies, Monash University, Clayton, VIC 3800, Australia
| | - Laurence Meagher
- Department of Materials Science and Engineering, Monash University, Clayton, Victoria, Australia; ARC Training Centre for Cell and Tissue Engineering Technologies, Monash University, Clayton, VIC 3800, Australia.
| |
Collapse
|
31
|
Zhang Y, Zouboulis CC, Xiao Z. Exosomes from adipose-derived stem cells activate sebocytes through the PI3K/AKT/SREBP-1 pathway to accelerate wound healing. Cell Tissue Res 2024; 396:329-342. [PMID: 38411945 PMCID: PMC11144157 DOI: 10.1007/s00441-024-03872-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: 05/29/2023] [Accepted: 01/25/2024] [Indexed: 02/28/2024]
Abstract
Sebocyte regeneration after injury is considered a key element of functional skin repair. Exosomes from adipose-derived stem cells (ADSCs-EXO) accelerate wound healing by promoting the proliferation of fibroblasts. However, the effects of ADSCs-EXO on sebocytes are largely unknown. In this study, the effects of ADSCs-EXO on sebocyte proliferation and migration were evaluated. The levels of phosphorylated AKT (p-AKT), AKT, sterol regulatory-element binding protein (SREBP), and perilipin-1 (PLIN-1) were detected with immunofluorescence, quantitative PCR, and western blot analysis. RNA-Seq was used to analyze the differential gene expression between the ADSCs-EXO group and the control group under anaerobic conditions. Lipogenesis was assessed with Nile red staining. In animal studies, full-thickness skin wounds in BALB/c mice were treated with gelatin methacrylate (GelMA) hydrogel-loaded sebocytes alone or in combination with ADSCs-EXO. Histopathological assessments of the wound tissues were performed Masson Trichrome staining, Immunohistochemical staining and so on. The phosphatidylinositol 3-kinase (PI3K)/AKT pathway blocker LY294002 inhibited the effects of ADSCs-EXO on p-AKT and sebocytes proliferation. ADSCs-EXO also regulated the expression of SREBP-1 and PLIN-1 through the PI3K/AKT pathway in an oxygen level-dependent manner. In BALB/c mice, ADSCs-EXO accelerated sebocyte-assisted wound healing and regeneration. These in vitro and in vivo results supported that ADSCs-EXO can promote the regeneration of fully functional skin after injury through the PI3K/AKT-dependent activation of sebocytes.
Collapse
Affiliation(s)
- Yingbo Zhang
- Department of Plastic and Aesthetic Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150081, People's Republic of China
| | - Christos C Zouboulis
- Departments of Dermatology, Venereology, Allergology and Immunology, Dessau Medical Center, Brandenburg Medical School Theodor Fontane and Faculty of Health Sciences Brandenburg, Dessau, Germany
| | - Zhibo Xiao
- Department of Plastic and Aesthetic Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150081, People's Republic of China.
| |
Collapse
|
32
|
Luo Y, Gao Y. Potential Role of Hydrogels in Stem Cell Culture and Hepatocyte Differentiation. NANO BIOMEDICINE AND ENGINEERING 2024; 16:188-202. [DOI: 10.26599/nbe.2024.9290055] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/21/2025]
|
33
|
Hu B, Qiao W, Cao Y, Fu X, Song J. A sono-responsive antibacterial nanosystem co-loaded with metformin and bone morphogenetic protein-2 for mitigation of inflammation and bone loss in experimental peri-implantitis. Front Bioeng Biotechnol 2024; 12:1410230. [PMID: 38854857 PMCID: PMC11157067 DOI: 10.3389/fbioe.2024.1410230] [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: 03/31/2024] [Accepted: 05/06/2024] [Indexed: 06/11/2024] Open
Abstract
Background Dental implants have become an increasingly popular option for replacing missing teeth, and the prevalence of peri-implantitis has also increased, which is expected to become a public health problem worldwide and cause high economic and health burdens. This scenario highlights the need for new therapeutic options to treat peri-implantitis. Methods In this study, we proposed a novel sono-responsive antibacterial nanosystem co-loaded with metformin (Met) and bone morphogenetic protein-2 (BMP-2) to promote efficacy in treating peri-implantitis. We introduced the zeolitic imidazolate framework-8 (ZIF-8) as a carrier for hematoporphyrin monomethyl ether (HMME) to enhance the antibacterial effect of sonodynamic antibacterial therapy and tested its reactive oxygen species (ROS) production efficiency and bactericidal effect in vitro. Afterward, HMME-loaded ZIF-8, BMP-2-loaded polylactic acid-glycolic acid (PLGA), and Met were incorporated into gelatin methacryloyl (GelMA) hydrogels to form HMME@ZIF-8/Met/BMP-2@PLGA/GelMA composite hydrogels, and the biocompatibility of which was determined in vitro and in vivo. A bacterial-induced peri-implantitis model in the maxilla of rats was established to detect the effects of the composite hydrogels with adjunctive use of ultrasound on regulating inflammation and promoting bone tissue repair in vivo. Results The results indicated that HMME@ZIF-8 with ultrasound stimulation demonstrated more better ROS production efficiency and antimicrobial efficacy. The composite hydrogels had good biocompatibility. Ultrasound-assisted application of the composite hydrogels reduced the release of the inflammatory factors IL-6 and TNF-α and reduced bone loss around the implant in rats with bacterial-induced peri-implantitis. Conclusion Our observations suggest that HMME@ZIF-8 may be a new good sonosensitizer material for sonodynamic antibacterial therapy. The use of HMME@ZIF-8/Met/BMP-2@PLGA/GelMA composite hydrogels in combination with ultrasound can provide a novel option for treating peri-implantitis in the future.
Collapse
Affiliation(s)
- Bo Hu
- College of Stomatology, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Oral Diseases, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Wang Qiao
- Department of Stomatology, Shapingba Hospital Affiliated to Chongqing University, Chongqing, China
| | - Yang Cao
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, Institute of Ultrasound Imaging, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaoming Fu
- College of Stomatology, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Oral Diseases, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Jinlin Song
- College of Stomatology, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Oral Diseases, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| |
Collapse
|
34
|
Mendoza-Cerezo L, Rodríguez-Rego JM, Macías-García A, Callejas-Marín A, Sánchez-Guardado L, Marcos-Romero AC. Three-Dimensional Bioprinting of GelMA Hydrogels with Culture Medium: Balancing Printability, Rheology and Cell Viability for Tissue Regeneration. Polymers (Basel) 2024; 16:1437. [PMID: 38794630 PMCID: PMC11124935 DOI: 10.3390/polym16101437] [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: 03/27/2024] [Revised: 04/14/2024] [Accepted: 05/10/2024] [Indexed: 05/26/2024] Open
Abstract
Three-dimensional extrusion bioprinting technology aims to become a fundamental tool for tissue regeneration using cell-loaded hydrogels. These biomaterials must have highly specific mechanical and biological properties that allow them to generate biosimilar structures by successive layering of material while maintaining cell viability. The rheological properties of hydrogels used as bioinks are critical to their printability. Correct printability of hydrogels allows the replication of biomimetic structures, which are of great use in medicine, tissue engineering and other fields of study that require the three-dimensional replication of different tissues. When bioprinting cell-loaded hydrogels, a small amount of culture medium can be added to ensure adequate survival, which can modify the rheological properties of the hydrogels. GelMA is a hydrogel used in bioprinting, with very interesting properties and rheological parameters that have been studied and defined for its basic formulation. However, the changes that occur in its rheological parameters and therefore in its printability, when it is mixed with the culture medium necessary to house the cells inside, are unknown. Therefore, in this work, a comparative study of GelMA 100% and GelMA in the proportions 3:1 (GelMA 75%) and 1:1 (GelMA 50%) with culture medium was carried out to determine the printability of the gel (using a device of our own invention), its main rheological parameters and its toxicity after the addition of the medium and to observe whether significant differences in cell viability occur. This raises the possibility of its use in regenerative medicine using a 3D extrusion bioprinter.
Collapse
Affiliation(s)
- Laura Mendoza-Cerezo
- Department of Graphic Expression, School of Industrial Engineering, University of Extremadura, Avenida de Elvas, s/n, 06006 Badajoz, Spain; (L.M.-C.); (A.C.M.-R.)
| | - Jesús M. Rodríguez-Rego
- Department of Graphic Expression, School of Industrial Engineering, University of Extremadura, Avenida de Elvas, s/n, 06006 Badajoz, Spain; (L.M.-C.); (A.C.M.-R.)
| | - Antonio Macías-García
- Department of Mechanical, Energy and Materials Engineering, School of Industrial Engineering, University of Extremadura, Avenida de Elvas, s/n, 06006 Badajoz, Spain;
| | - Antuca Callejas-Marín
- Department of Anatomy, Cell Biology and Zoology, Faculty of Science, University of Extremadura, Avenida de Elvas, s/n, 06006 Badajoz, Spain; (A.C.-M.); (L.S.-G.)
| | - Luís Sánchez-Guardado
- Department of Anatomy, Cell Biology and Zoology, Faculty of Science, University of Extremadura, Avenida de Elvas, s/n, 06006 Badajoz, Spain; (A.C.-M.); (L.S.-G.)
| | - Alfonso C. Marcos-Romero
- Department of Graphic Expression, School of Industrial Engineering, University of Extremadura, Avenida de Elvas, s/n, 06006 Badajoz, Spain; (L.M.-C.); (A.C.M.-R.)
| |
Collapse
|
35
|
Zhang B, Bi Y, Wang K, Guo X, Liu Z, Li J, Wu M. Stem Cell-Derived Extracellular Vesicles: Promising Therapeutic Opportunities for Diabetic Wound Healing. Int J Nanomedicine 2024; 19:4357-4375. [PMID: 38774027 PMCID: PMC11108067 DOI: 10.2147/ijn.s461342] [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: 01/25/2024] [Accepted: 05/10/2024] [Indexed: 05/24/2024] Open
Abstract
Wound healing is a sophisticated and orderly process of cellular interactions in which the body restores tissue architecture and functionality following injury. Healing of chronic diabetic wounds is difficult due to impaired blood circulation, a reduced immune response, and disrupted cellular repair mechanisms, which are often associated with diabetes. Stem cell-derived extracellular vesicles (SC-EVs) hold the regenerative potential, encapsulating a diverse cargo of proteins, RNAs, and cytokines, presenting a safe, bioactivity, and less ethical issues than other treatments. SC-EVs orchestrate multiple regenerative processes by modulating cellular communication, increasing angiogenesis, and promoting the recruitment and differentiation of progenitor cells, thereby potentiating the reparative milieu for diabetic wound healing. Therefore, this review investigated the effects and mechanisms of EVs from various stem cells in diabetic wound healing, as well as their limitations and challenges. Continued exploration of SC-EVs has the potential to revolutionize diabetic wound care.
Collapse
Affiliation(s)
- Boyu Zhang
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People’s Republic of China
| | - Yajun Bi
- Department of Pediatrics, Dalian Municipal Women and Children’s Medical Center (Group), Dalian Medical University, Dalian, Liaoning Province, 116011, People’s Republic of China
| | - Kang Wang
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People’s Republic of China
| | - Xingjun Guo
- Department of Biliary-Pancreatic Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, People’s Republic of China
| | - Zeming Liu
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People’s Republic of China
| | - Jia Li
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People’s Republic of China
| | - Min Wu
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People’s Republic of China
| |
Collapse
|
36
|
Bupphathong S, Lim J, Fang HW, Tao HY, Yeh CE, Ku TA, Huang W, Kuo TY, Lin CH. Enhanced Vascular-like Network Formation of Encapsulated HUVECs and ADSCs Coculture in Growth Factors Conjugated GelMA Hydrogels. ACS Biomater Sci Eng 2024; 10:3306-3315. [PMID: 38634810 PMCID: PMC11094682 DOI: 10.1021/acsbiomaterials.4c00465] [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: 03/09/2024] [Revised: 04/08/2024] [Accepted: 04/08/2024] [Indexed: 04/19/2024]
Abstract
Tissue engineering primarily aimed to alleviate the insufficiency of organ donations worldwide. Nonetheless, the survival of the engineered tissue is often compromised due to the complexity of the natural organ architectures, especially the vascular system inside the organ, which allows food-waste transfer. Thus, vascularization within the engineered tissue is of paramount importance. A critical aspect of this endeavor is the ability to replicate the intricacies of the extracellular matrix and promote the formation of functional vascular networks within engineered constructs. In this study, human adipose-derived stem cells (hADSCs) and human umbilical vein endothelial cells (HUVECs) were cocultured in different types of gelatin methacrylate (GelMA). In brief, pro-angiogenic signaling growth factors (GFs), vascular endothelial growth factor (VEGF165) and basic fibroblast growth factor (bFGF), were conjugated onto GelMA via an EDC/NHS coupling reaction. The GelMA hydrogels conjugated with VEGF165 (GelMA@VEGF165) and bFGF (GelMA@bFGF) showed marginal changes in the chemical and physical characteristics of the GelMA hydrogels. Moreover, the conjugation of these growth factors demonstrated improved cell viability and cell proliferation within the hydrogel construct. Additionally, vascular-like network formation was observed predominantly on GelMA@GrowthFactor (GelMA@GF) hydrogels, particularly on GelMA@bFGF. This study suggests that growth factor-conjugated GelMA hydrogels would be a promising biomaterial for 3D vascular tissue engineering.
Collapse
Affiliation(s)
- Sasinan Bupphathong
- Graduate
Institute of Nanomedicine and Medical Engineering, College of Biomedical
Engineering, Taipei Medical University, Taipei 110, Taiwan
- High-Value
Biomaterials Research and Commercialization Center, National Taipei University of Technology, Taipei 10608, Taiwan
| | - Joshua Lim
- Graduate
Institute of Nanomedicine and Medical Engineering, College of Biomedical
Engineering, Taipei Medical University, Taipei 110, Taiwan
| | - Hsu-Wei Fang
- High-Value
Biomaterials Research and Commercialization Center, National Taipei University of Technology, Taipei 10608, Taiwan
- Department
of Chemical Engineering and Biotechnology, National Taipei University of Technology, Taipei 10608, Taiwan
- Institute
of Biomedical Engineering and Nanomedicine, National Health Research Institutes, Zhunan 35053, Taiwan
| | - Hsuan-Ya Tao
- Graduate
Institute of Nanomedicine and Medical Engineering, College of Biomedical
Engineering, Taipei Medical University, Taipei 110, Taiwan
| | - Chen-En Yeh
- School
of Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 110, Taiwan
| | - Tian-An Ku
- School
of Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 110, Taiwan
| | - Wei Huang
- Department
of Orthodontics, Rutgers School of Dental
Medicine, Newark, New Jersey 07103, United States
| | - Ting-Yu Kuo
- School
of Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 110, Taiwan
| | - Chih-Hsin Lin
- Graduate
Institute of Nanomedicine and Medical Engineering, College of Biomedical
Engineering, Taipei Medical University, Taipei 110, Taiwan
| |
Collapse
|
37
|
Sun J, Xie X, Song Y, Sun T, Liu X, Yuan H, Shen C. Selenomethionine in gelatin methacryloyl hydrogels: Modulating ferroptosis to attenuate skin aging. Bioact Mater 2024; 35:495-516. [PMID: 38404642 PMCID: PMC10885793 DOI: 10.1016/j.bioactmat.2024.02.013] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 02/08/2024] [Accepted: 02/09/2024] [Indexed: 02/27/2024] Open
Abstract
During skin aging, the degeneration of epidermal stem cells (EpiSCs) leads to diminished wound healing capabilities and epidermal disintegration. This study tackles this issue through a comprehensive analysis combining transcriptomics and untargeted metabolomics, revealing age-dependent alterations in the Gpx gene family and arachidonic acid (AA) metabolic networks, resulting in enhanced ferroptosis. Selenomethionine (Se-Met) could enhance GPX4 expression, thereby assisting EpiSCs in countering AA-induced mitochondrial damage and ferroptosis. Additionally, Se-Met demonstrates antioxidative characteristics and extensive ultraviolet absorption. For the sustained and controllable release of Se-Met, it was covalently grafted to UV-responsive GelMA hydrogels via AC-PEG-NHS tethers. The Se-Met@GelMA hydrogel effectively accelerated wound healing in a chronological aging mice model, by inhibiting lipid peroxidation and ferroptosis with augmented GPX4 expression. Moreover, in a photoaging model, this hydrogel significantly mitigated inflammatory responses, extracellular matrix remodeling, and ferroptosis in UV-exposed mice. These characteristics render Se-Met@GelMA hydrogel valuable in practical clinical applications.
Collapse
Affiliation(s)
- Jiachen Sun
- Department of Burns and Plastic Surgery, Fourth Medical Center of Chinese PLA General Hospital, Beijing, 100048, China
| | - Xiaoye Xie
- Department of Burns and Plastic Surgery, Fourth Medical Center of Chinese PLA General Hospital, Beijing, 100048, China
| | - Yaoyao Song
- Department of Burns and Plastic Surgery, Fourth Medical Center of Chinese PLA General Hospital, Beijing, 100048, China
| | - Tianjun Sun
- Department of Burns and Plastic Surgery, Fourth Medical Center of Chinese PLA General Hospital, Beijing, 100048, China
| | - Xinzhu Liu
- Department of Burns and Plastic Surgery, Fourth Medical Center of Chinese PLA General Hospital, Beijing, 100048, China
| | - Huageng Yuan
- Department of Burns and Plastic Surgery, Fourth Medical Center of Chinese PLA General Hospital, Beijing, 100048, China
| | - Chuanan Shen
- Department of Burns and Plastic Surgery, Fourth Medical Center of Chinese PLA General Hospital, Beijing, 100048, China
| |
Collapse
|
38
|
Cao H, Wang J, Hao Z, Zhao D. Gelatin-based biomaterials and gelatin as an additive for chronic wound repair. Front Pharmacol 2024; 15:1398939. [PMID: 38751781 PMCID: PMC11094280 DOI: 10.3389/fphar.2024.1398939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 04/15/2024] [Indexed: 05/18/2024] Open
Abstract
Disturbing or disrupting the regular healing process of a skin wound may result in its progression to a chronic state. Chronic wounds often lead to increased infection because of their long healing time, malnutrition, and insufficient oxygen flow, subsequently affecting wound progression. Gelatin-the main structure of natural collagen-is widely used in biomedical fields because of its low cost, wide availability, biocompatibility, and degradability. However, gelatin may exhibit diverse tailored physical properties and poor antibacterial activity. Research on gelatin-based biomaterials has identified the challenges of improving gelatin's poor antibacterial properties and low mechanical properties. In chronic wounds, gelatin-based biomaterials can promote wound hemostasis, enhance peri-wound antibacterial and anti-inflammatory properties, and promote vascular and epithelial cell regeneration. In this article, we first introduce the natural process of wound healing. Second, we present the role of gelatin-based biomaterials and gelatin as an additive in wound healing. Finally, we present the future implications of gelatin-based biomaterials.
Collapse
Affiliation(s)
- Hongwei Cao
- Department of Otorhinolaryngology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Jingren Wang
- Department of Prosthodontics, Affiliated Stomatological Hospital of China Medical University, Shenyang, China
| | - Zhanying Hao
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Danyang Zhao
- Department of emergency Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| |
Collapse
|
39
|
Wang H, Wan J, Zhang Z, Hou R. Recent advances on 3D-bioprinted gelatin methacrylate hydrogels for tissue engineering in wound healing: A review of current applications and future prospects. Int Wound J 2024; 21:e14533. [PMID: 38069620 PMCID: PMC10961039 DOI: 10.1111/iwj.14533] [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: 10/14/2023] [Revised: 11/16/2023] [Accepted: 11/18/2023] [Indexed: 03/25/2024] Open
Abstract
Advancements in 3D bioprinting, particularly the use of gelatin methacrylate (GelMA) hydrogels, are ushering in a transformative era in regenerative medicine and tissue engineering. This review highlights the pivotal role of GelMA hydrogels in wound healing and skin regeneration. Its biocompatibility, tunable mechanical properties and support for cellular proliferation make it a promising candidate for bioactive dressings and scaffolds. Challenges remain in optimizing GelMA hydrogels for clinical use, including scalability of 3D bioprinting techniques, durability under physiological conditions and the development of advanced bioinks. The review covers GelMA's applications from enhancing wound dressings, promoting angiogenesis and facilitating tissue regeneration to addressing microbial infections and diabetic wound healing. Preclinical studies underscore GelMA's potential in tissue healing and the need for further research for real-world applications. The future of GelMA hydrogels lies in overcoming these challenges through multidisciplinary collaboration, advancing manufacturing techniques and embracing personalized medicine paradigms.
Collapse
Affiliation(s)
- Hongyu Wang
- Department of OrthopedicsSuzhou Medical College of Soochow UniversitySuzhouChina
| | - Jiaming Wan
- Department of OrthopedicsYangzhou University Medical CollegeYangzhouChina
| | - Zhiqiang Zhang
- Department of OrthopedicsSuzhou Medical College of Soochow UniversitySuzhouChina
| | - Ruixing Hou
- Department of OrthopedicsSuzhou Medical College of Soochow UniversitySuzhouChina
- Department of Trauma OrthopedicsSuzhou Ruihua Orthopedic HospitalSuzhouChina
| |
Collapse
|
40
|
Lu T, Liu Y, Huang X, Sun S, Xu H, Jin A, Wang X, Gao X, Liu J, Zhu Y, Dai Q, Wang C, Lin K, Jiang L. Early-Responsive Immunoregulation Therapy Improved Microenvironment for Bone Regeneration Via Engineered Extracellular Vesicles. Adv Healthc Mater 2024; 13:e2303681. [PMID: 38054523 DOI: 10.1002/adhm.202303681] [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: 10/25/2023] [Indexed: 12/07/2023]
Abstract
Overactivated inflammatory reactions hinder the bone regeneration process. Timely transformation of microenvironment from pro-inflammatory to anti-inflammatory after acute immune response is favorable for osteogenesis. Macrophages play an important role in the immune response to inflammation. Therefore, this study adopts TIM3 high expression extracellular vesicles (EVs) with immunosuppressive function to reshape the early immune microenvironment of bone injury, mainly by targeting macrophages. These EVs can be phagocytosed by macrophages, thereby increasing the infiltration of TIM3-positive macrophages (TIM3+ macrophages) and M2 subtypes. The TIM3+ macrophage group has some characteristics of M2 macrophages and secretes cytokines, such as IL-10 and TGF-β1 to regulate inflammation. TIM3, which is highly expressed in the engineered EVs, mediates the release of anti-inflammatory cytokines by inhibiting the p38/MAPK pathway and promotes osseointegration by activating the Bmp2 promoter to enhance macrophage BMP2 secretion. After evenly loading the engineered EVs into the hydrogel, the continuous and slow release of EVsTIM3OE recruits more anti-inflammatory macrophages during the early stages of bone defect repair, regulating the immune microenvironment and eliminating the adverse effects of excessive inflammation. In summary, this study provides a new strategy for the treatment of refractory wounds through early inflammation control.
Collapse
Affiliation(s)
- Tingwei Lu
- Center of Craniofacial Orthodontics, Department of Oral and Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, 200011, China
| | - Yuanqi Liu
- Center of Craniofacial Orthodontics, Department of Oral and Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, 200011, China
| | - Xiangru Huang
- Center of Craniofacial Orthodontics, Department of Oral and Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, 200011, China
| | - Siyuan Sun
- Center of Craniofacial Orthodontics, Department of Oral and Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, 200011, China
| | - Hongyuan Xu
- Center of Craniofacial Orthodontics, Department of Oral and Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, 200011, China
| | - Anting Jin
- Center of Craniofacial Orthodontics, Department of Oral and Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, 200011, China
| | - Xinyu Wang
- Center of Craniofacial Orthodontics, Department of Oral and Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, 200011, China
| | - Xin Gao
- Center of Craniofacial Orthodontics, Department of Oral and Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, 200011, China
| | - Jingyi Liu
- Center of Craniofacial Orthodontics, Department of Oral and Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, 200011, China
| | - Yanfei Zhu
- Center of Craniofacial Orthodontics, Department of Oral and Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, 200011, China
| | - Qinggang Dai
- The 2nd Dental Center, Ninth People's Hospital, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, 201999, China
| | - Chao Wang
- Department of Obstetrics & Gynecology, Obstetrics & Gynecology Hospital of Fudan University, Shanghai, 200433, China
| | - Kaili Lin
- Center of Craniofacial Orthodontics, Department of Oral and Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, 200011, China
| | - Lingyong Jiang
- Center of Craniofacial Orthodontics, Department of Oral and Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, 200011, China
| |
Collapse
|
41
|
He Y, Cen Y, Tian M. Immunomodulatory hydrogels for skin wound healing: cellular targets and design strategy. J Mater Chem B 2024; 12:2435-2458. [PMID: 38284157 DOI: 10.1039/d3tb02626d] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2024]
Abstract
Skin wounds significantly impact the global health care system and represent a significant burden on the economy and society due to their complicated dynamic healing processes, wherein a series of immune events are required to coordinate normal and sequential healing phases, involving multiple immunoregulatory cells such as neutrophils, macrophages, keratinocytes, and fibroblasts, since dysfunction of these cells may impede skin wound healing presenting persisting inflammation, impaired vascularization, and excessive collagen deposition. Therefore, cellular target-based immunomodulation is promising to promote wound healing as cells are the smallest unit of life in immune response. Recently, immunomodulatory hydrogels have become an attractive avenue to promote skin wound healing. However, a detailed and comprehensive review of cellular targets and related hydrogel design strategies remains lacking. In this review, the roles of the main immunoregulatory cells participating in skin wound healing are first discussed, and then we highlight the cellular targets and state-of-the-art design strategies for immunomodulatory hydrogels based on immunoregulatory cells that cover defect, infected, diabetic, burn and tumor wounds and related scar healing. Finally, we discuss the barriers that need to be addressed and future prospects to boost the development and prosperity of immunomodulatory hydrogels.
Collapse
Affiliation(s)
- Yinhai He
- Department of Plastic and Burn Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Ying Cen
- Department of Plastic and Burn Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Meng Tian
- Department of Neurosurgery and Neurosurgery Research Laboratory, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
42
|
Jin M, Yi X, Zhu X, Hu W, Wang S, Chen Q, Yang W, Li Y, Li S, Peng Q, Pan M, Gao Y, Xu S, Zhang Y, Zhou S. Schisandrin B promotes hepatic differentiation from human umbilical cord mesenchymal stem cells. iScience 2024; 27:108912. [PMID: 38323006 PMCID: PMC10844828 DOI: 10.1016/j.isci.2024.108912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 10/30/2023] [Accepted: 01/11/2024] [Indexed: 02/08/2024] Open
Abstract
Human umbilical cord mesenchymal stem cells (UC-MSCs)-derived hepatocyte-like cells (HLCs) have shown great promise in the treatment of liver diseases. However, most current induction protocols yield hepatocyte-like cells with limited function as compared with primary hepatocytes. Schisandrin B (Sch B) is one of the main components of Schisandra chinensis, which can prevent fibrosis progression and promote liver cell regeneration. Herein, we investigated the effects of Sch B on hepatic differentiation of UC-MSCs. We found that treatment with 10 μM Sch B from the second stage of the differentiation process increased hepatic marker levels and hepatic function. Additionally, RNA-seq analysis revealed that Sch B promoted hepatic differentiation via activating the JAK2/STAT3 pathway. When transplanted HLCs into mice with CCL4-induced liver fibrosis, Sch B-treated HLCs exhibited significant therapeutic effects. This study provides an optimized hepatic differentiation protocol for UC-MSCs based on Sch B, yielding functioning cells for liver disease treatment.
Collapse
Affiliation(s)
- Meixian Jin
- Department of Anesthesiology, Zhujiang Hospital, Southern Medical University, Guangzhou 510000, China
| | - Xiao Yi
- Department of Gynecology, Zhujiang Hospital, Southern Medical University, Guangzhou 510000, China
| | - Xiaojuan Zhu
- Department of Anesthesiology, First People’s Hospital of Kashi, Kashi 844000, China
| | - Wei Hu
- Department of Anesthesiology, Zhujiang Hospital, Southern Medical University, Guangzhou 510000, China
| | - Simin Wang
- Department of Anesthesiology, Zhujiang Hospital, Southern Medical University, Guangzhou 510000, China
| | - Qi Chen
- Department of Anesthesiology, Zhujiang Hospital, Southern Medical University, Guangzhou 510000, China
| | - Wanren Yang
- General Surgery Center, Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center for Artificial Organ and Tissue Engineering, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Institute of Regenerative Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510000, China
| | - Yang Li
- General Surgery Center, Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center for Artificial Organ and Tissue Engineering, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Institute of Regenerative Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510000, China
| | - Shao Li
- General Surgery Center, Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center for Artificial Organ and Tissue Engineering, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Institute of Regenerative Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510000, China
| | - Qing Peng
- General Surgery Center, Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center for Artificial Organ and Tissue Engineering, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Institute of Regenerative Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510000, China
| | - Mingxin Pan
- General Surgery Center, Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center for Artificial Organ and Tissue Engineering, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Institute of Regenerative Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510000, China
| | - Yi Gao
- General Surgery Center, Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center for Artificial Organ and Tissue Engineering, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Institute of Regenerative Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510000, China
| | - Shiyuan Xu
- Department of Anesthesiology, Zhujiang Hospital, Southern Medical University, Guangzhou 510000, China
| | - Ying Zhang
- General Surgery Center, Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center for Artificial Organ and Tissue Engineering, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Institute of Regenerative Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510000, China
| | - Shuqin Zhou
- Department of Anesthesiology, Zhujiang Hospital, Southern Medical University, Guangzhou 510000, China
- Anesthesiology Department of The Second Affiliated Hospital, School of Medicine, The Chinese University of Hong Kong, Shenzhen & Longgang District People’s Hospital of Shenzhen, Shenzhen 518172, China
| |
Collapse
|
43
|
Bai X, Wang R, Hu X, Dai Q, Guo J, Cao T, Du W, Cheng Y, Xia S, Wang D, Yang L, Teng L, Chen D, Liu Y. Two-Dimensional Biodegradable Black Phosphorus Nanosheets Promote Large Full-Thickness Wound Healing through In Situ Regeneration Therapy. ACS NANO 2024; 18:3553-3574. [PMID: 38226901 PMCID: PMC10832999 DOI: 10.1021/acsnano.3c11177] [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: 11/11/2023] [Revised: 12/16/2023] [Accepted: 12/18/2023] [Indexed: 01/17/2024]
Abstract
Large full-thickness skin lesions have been one of the most challenging clinical problems in plastic surgery repair and reconstruction. To achieve in situ skin regeneration and perfect clinical outcomes, we must address two significant obstacles: angiogenesis deficiency and inflammatory dysfunction. Recently, black phosphorus has shown great promise in wound healing. However, few studies have explored the bio-effects of BP to promote in situ skin regeneration based on its nanoproperties. Here, to investigate whether black phosphorus nanosheets have positive bio-effects on in situ skin repair, we verified black phosphorus nanosheets' positive effects on angiogenic and anti-inflammatory abilities in vitro. Next, the in vivo evaluation performed on the rat large full-thickness excisional wound splinting model more comprehensively showed that the positive bio-effects of black phosphorus nanosheets are multilevel in wound healing, which can effectively enhance anti-inflammatory ability, angiogenesis, collagen deposition, and skin re-epithelialization. Then, multiomics analysis was performed to explore further the mechanism of black phosphorus nanosheets' regulation of endothelial cells in depth. Molecular mechanistically, black phosphorus nanosheets activated the JAK-STAT-OAS signaling pathway to promote cellular function and mitochondrial energy metabolism in endothelial cells. This study can provide a theoretical basis for applying two-dimensional black phosphorus nanosheets as nanomedicine to achieve in situ tissue regeneration in complex human pathological microenvironments, guiding the subsequent optimization of black phosphorus.
Collapse
Affiliation(s)
- Xueshan Bai
- Cranio-Maxillo-Facial
Surgery Department, Plastic Surgery Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100144, China
| | - Renxian Wang
- Laboratory
of Bone Tissue Engineering, Beijing Laboratory of Biomedical Materials,
National Center for Orthopaedics, Beijing Research Institute of Traumatology
and Orthopaedics, Beijing Jishuitan Hospital, Capital Medical University, Beijing 100035, China
- JST
sarcopenia Research Centre, National Center for Orthopaedics, Beijing
Research Institute of Traumatology and Orthopaedics, Beijing Jishuitan
Hospital, Capital Medical University, Beijing 100035, China
| | - Xiaohua Hu
- Department
of Burns and Plastic Surgery, Beijing Jishuitan Hospital, Capital Medical University, Beijing 100035, China
| | - Qiang Dai
- Department
of Burns and Plastic Surgery, Beijing Jishuitan Hospital, Capital Medical University, Beijing 100035, China
| | - Jianxun Guo
- Laboratory
of Bone Tissue Engineering, Beijing Laboratory of Biomedical Materials,
National Center for Orthopaedics, Beijing Research Institute of Traumatology
and Orthopaedics, Beijing Jishuitan Hospital, Capital Medical University, Beijing 100035, China
| | - Tongyu Cao
- Department
of Burns and Plastic Surgery, Beijing Jishuitan Hospital, Capital Medical University, Beijing 100035, China
| | - Weili Du
- Department
of Burns and Plastic Surgery, Beijing Jishuitan Hospital, Capital Medical University, Beijing 100035, China
| | - Yuning Cheng
- Laboratory
of Bone Tissue Engineering, Beijing Laboratory of Biomedical Materials,
National Center for Orthopaedics, Beijing Research Institute of Traumatology
and Orthopaedics, Beijing Jishuitan Hospital, Capital Medical University, Beijing 100035, China
| | - Songxia Xia
- Cranio-Maxillo-Facial
Surgery Department, Plastic Surgery Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100144, China
| | - Dingding Wang
- JST
sarcopenia Research Centre, National Center for Orthopaedics, Beijing
Research Institute of Traumatology and Orthopaedics, Beijing Jishuitan
Hospital, Capital Medical University, Beijing 100035, China
| | - Liya Yang
- Cranio-Maxillo-Facial
Surgery Department, Plastic Surgery Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100144, China
| | - Li Teng
- Cranio-Maxillo-Facial
Surgery Department, Plastic Surgery Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100144, China
| | - Dafu Chen
- Laboratory
of Bone Tissue Engineering, Beijing Laboratory of Biomedical Materials,
National Center for Orthopaedics, Beijing Research Institute of Traumatology
and Orthopaedics, Beijing Jishuitan Hospital, Capital Medical University, Beijing 100035, China
| | - Yajun Liu
- JST
sarcopenia Research Centre, National Center for Orthopaedics, Beijing
Research Institute of Traumatology and Orthopaedics, Beijing Jishuitan
Hospital, Capital Medical University, Beijing 100035, China
- Department
of Spine Surgery, Beijing Jishuitan Hospital, National Center for
Orthopaedics, Beijing Jishuitan Hospital, Capital Medical University, Beijing 100035, China
| |
Collapse
|
44
|
Nakipoglu M, Özkabadayı Y, Karahan S, Tezcaner A. Bilayer wound dressing composed of asymmetric polycaprolactone membrane and chitosan-carrageenan hydrogel incorporating storax balsam. Int J Biol Macromol 2024; 254:128020. [PMID: 37956814 DOI: 10.1016/j.ijbiomac.2023.128020] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 11/08/2023] [Accepted: 11/09/2023] [Indexed: 11/15/2023]
Abstract
A comprehensive approach is needed to develop multifunctional wound dressing that is simple yet efficient. In this work, Liquidambar orientalis Mill. storax loaded hydroxyethyl chitosan (HECS)-carrageenan (kC) based hydrogel (HECS-kC) and polydopamine coated asymmetric polycaprolactone membrane (PCL-DOP) were used to develop a multifunctional and modular bilayer wound dressing. Asymmetric PCL-DOP membrane was prepared by non-solvent induced phase separation (NIPS) followed by polydopamine coating and demonstrated an excellent barrier against bacteria while allowing permeability for 5.45 ppm dissolved‑oxygen and 2130 g/m2 water vapor transmission in 24 h in addition to 805 kPa tensile strength. Storax loaded HECS-kC hydrogel, on the other hand, demonstrated a pH-responsive degradation and swelling to provide necessary conditions to facilitate wound healing. The hydrogels showed stretchability above 140 %, mild adhesive strength on sheep skin and PCL-DOP membrane, while the storax incorporation enhanced antibacterial and antioxidant activity. Furthermore, rat full-thickness skin defect model showed that the developed bilayer wound dressing could significantly facilitate wound healing compared to Tegaderm™ and control groups. This study shows that the bilayered wound dressing has the potential to be used as a simple and effective wound care system.
Collapse
Affiliation(s)
- Mustafa Nakipoglu
- Department of Biotechnology, Middle East Technical University, Ankara 06800, Turkey; Department of Molecular Biology and Genetics, Bartin University, Bartin 74100, Turkey.
| | - Yasin Özkabadayı
- Department of Histology, Kırıkkale University, Kırıkkale 71450, Turkey.
| | - Siyami Karahan
- Department of Histology, Kırıkkale University, Kırıkkale 71450, Turkey.
| | - Ayşen Tezcaner
- Department of Biotechnology, Middle East Technical University, Ankara 06800, Turkey; Department of Engineering Sciences, Middle East Technical University, Ankara 06800, Turkey.
| |
Collapse
|
45
|
Zhu Y, Chen J, Liu H, Zhang W. Photo-cross-linked Hydrogels for Cartilage and Osteochondral Repair. ACS Biomater Sci Eng 2023; 9:6567-6585. [PMID: 37956022 DOI: 10.1021/acsbiomaterials.3c01132] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Photo-cross-linked hydrogels, which respond to light and induce structural or morphological transitions, form a microenvironment that mimics the extracellular matrix of native tissue. In the last decades, photo-cross-linked hydrogels have been widely used in cartilage and osteochondral tissue engineering due to their good biocompatibility, ease of fabrication, rapid in situ gel-forming ability, and tunable mechanical and degradable properties. In this review, we systemically summarize the different types and physicochemical properties of photo-cross-linked hydrogels (including the materials and photoinitiators) and explore the biological properties modulated through the incorporation of additives, including cells, biomolecules, genes, and nanomaterials, into photo-cross-linked hydrogels. Subsequently, we compile the applications of photo-cross-linked hydrogels with a specific focus on cartilage and osteochondral repair. Finally, current limitations and future perspectives of photo-cross-linked hydrogels are also discussed.
Collapse
Affiliation(s)
- Yue Zhu
- School of Medicine, Southeast University, 210009 Nanjing, China
| | - Jialin Chen
- School of Medicine, Southeast University, 210009 Nanjing, China
- Jiangsu Key Laboratory for Biomaterials and Devices, Southeast University, 210096 Nanjing, China
- China Orthopedic Regenerative Medicine Group (CORMed), 310058 Hangzhou, China
| | - Haoyang Liu
- School of Medicine, Southeast University, 210009 Nanjing, China
| | - Wei Zhang
- School of Medicine, Southeast University, 210009 Nanjing, China
- Jiangsu Key Laboratory for Biomaterials and Devices, Southeast University, 210096 Nanjing, China
- China Orthopedic Regenerative Medicine Group (CORMed), 310058 Hangzhou, China
| |
Collapse
|
46
|
Ai Y, Dai F, Li W, Xu F, Yang H, Wu J, Yang K, Li L, Ai F, Song L. Photo-crosslinked bioactive BG/BMSCs@GelMA hydrogels for bone-defect repairs. Mater Today Bio 2023; 23:100882. [PMID: 38161508 PMCID: PMC10755535 DOI: 10.1016/j.mtbio.2023.100882] [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: 09/02/2023] [Revised: 11/05/2023] [Accepted: 11/22/2023] [Indexed: 01/03/2024] Open
Abstract
The clinical treatments of bone defects remain a challenge. Hydrogels containing bone marrow mesenchymal stem cells (BMSCs) are extensively used to bone regeneration because of excellent biocompatibility and hydrophilicity. However, the insufficient osteo-induction capacity of the BMSC-loaded hydrogels limits their clinical applications. In this study, bio-active glass (BG) and BMSCs were combined with gelatin methacryloyl (GelMA) to fabricate composite hydrogels via photo-crosslinking, and the regulation of bone regeneration was investigated. In vitro experiments showed that the BG/BMSCs@GelMA hydrogel had excellent cytocompatibility and promoted osteogenic differentiation in BMSCs. Furthermore, the BG/BMSCs@GelMA hydrogel was injected into critical-sized calvarial defects, and the results further confirmed its excellent angiogenetic and bone regeneration capacity. In addition, BG/BMSCs@GelMA promoted the polarization of macrophages towards the M2 phenotype. In summary, this novel composite hydrogel demonstrated remarkable potential for application in bone regeneration due to its immunomodulatory, excellent angiogenetic as well as osteo-induction capacity.
Collapse
Affiliation(s)
- Yufeng Ai
- Center of Stomatology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 33006, China
- The Institute of Periodontal Disease, Nanchang University, Nanchang, Jiangxi, 33006, China
- JXHC Key Laboratory of Periodontology (The Second Affiliated Hospital of Nanchang University), Nanchang, Jiangxi, 33006, China
- The Second Clinical Medical School, Nanchang University, Nanchang, Jiangxi, 33006, China
| | - Fang Dai
- Center of Stomatology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 33006, China
- The Institute of Periodontal Disease, Nanchang University, Nanchang, Jiangxi, 33006, China
- JXHC Key Laboratory of Periodontology (The Second Affiliated Hospital of Nanchang University), Nanchang, Jiangxi, 33006, China
| | - Wenfeng Li
- Center of Stomatology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 33006, China
- The Institute of Periodontal Disease, Nanchang University, Nanchang, Jiangxi, 33006, China
- JXHC Key Laboratory of Periodontology (The Second Affiliated Hospital of Nanchang University), Nanchang, Jiangxi, 33006, China
| | - Fancheng Xu
- Center of Stomatology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 33006, China
- The Institute of Periodontal Disease, Nanchang University, Nanchang, Jiangxi, 33006, China
- JXHC Key Laboratory of Periodontology (The Second Affiliated Hospital of Nanchang University), Nanchang, Jiangxi, 33006, China
- The Second Clinical Medical School, Nanchang University, Nanchang, Jiangxi, 33006, China
| | - Hanwen Yang
- The Second Clinical Medical School, Nanchang University, Nanchang, Jiangxi, 33006, China
| | - Jianxin Wu
- Center of Stomatology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 33006, China
- The Institute of Periodontal Disease, Nanchang University, Nanchang, Jiangxi, 33006, China
- JXHC Key Laboratory of Periodontology (The Second Affiliated Hospital of Nanchang University), Nanchang, Jiangxi, 33006, China
- The Second Clinical Medical School, Nanchang University, Nanchang, Jiangxi, 33006, China
| | - Kaiqiang Yang
- Center of Stomatology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 33006, China
- The Institute of Periodontal Disease, Nanchang University, Nanchang, Jiangxi, 33006, China
- JXHC Key Laboratory of Periodontology (The Second Affiliated Hospital of Nanchang University), Nanchang, Jiangxi, 33006, China
- The Second Clinical Medical School, Nanchang University, Nanchang, Jiangxi, 33006, China
| | - Li Li
- Center of Stomatology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 33006, China
- The Institute of Periodontal Disease, Nanchang University, Nanchang, Jiangxi, 33006, China
- JXHC Key Laboratory of Periodontology (The Second Affiliated Hospital of Nanchang University), Nanchang, Jiangxi, 33006, China
- The Second Clinical Medical School, Nanchang University, Nanchang, Jiangxi, 33006, China
| | - Fanrong Ai
- School of Advanced Manufacturing, Nanchang University, Nanchang, Jiangxi, 33006, China
| | - Li Song
- Center of Stomatology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 33006, China
- The Institute of Periodontal Disease, Nanchang University, Nanchang, Jiangxi, 33006, China
- JXHC Key Laboratory of Periodontology (The Second Affiliated Hospital of Nanchang University), Nanchang, Jiangxi, 33006, China
| |
Collapse
|
47
|
Mahjoor M, Fakouri A, Farokhi S, Nazari H, Afkhami H, Heidari F. Regenerative potential of mesenchymal stromal cells in wound healing: unveiling the influence of normoxic and hypoxic environments. Front Cell Dev Biol 2023; 11:1245872. [PMID: 37900276 PMCID: PMC10603205 DOI: 10.3389/fcell.2023.1245872] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 08/11/2023] [Indexed: 10/31/2023] Open
Abstract
The innate and adaptive immune systems rely on the skin for various purposes, serving as the primary defense against harmful environmental elements. However, skin lesions may lead to undesirable consequences such as scarring, accelerated skin aging, functional impairment, and psychological effects over time. The rising popularity of mesenchymal stromal cells (MSCs) for skin wound treatment is due to their potential as a promising therapeutic option. MSCs offer advantages in terms of differentiation capacity, accessibility, low immunogenicity, and their central role in natural wound-healing processes. To accelerate the healing process, MSCs promote cell migration, angiogenesis, epithelialization, and granulation tissue development. Oxygen plays a critical role in the formation and expansion of mammalian cells. The term "normoxia" refers to the usual oxygen levels, defined at 20.21 percent oxygen (160 mm of mercury), while "hypoxia" denotes oxygen levels of 2.91 percent or less. Notably, the ambient O2 content (20%) in the lab significantly differs from the 2%-9% O2 concentration in their natural habitat. Oxygen regulation of hypoxia-inducible factor-1 (HIF-1) mediated expression of multiple genes plays a crucial role in sustaining stem cell destiny concerning proliferation and differentiation. This study aims to elucidate the impact of normoxia and hypoxia on MSC biology and draw comparisons between the two. The findings suggest that expanding MSC-based regenerative treatments in a hypoxic environment can enhance their growth kinetics, genetic stability, and expression of chemokine receptors, ultimately increasing their effectiveness.
Collapse
Affiliation(s)
- Mohamad Mahjoor
- Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran
- Department of Immunology, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Arshia Fakouri
- Student Research Committee, USERN Office, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Simin Farokhi
- Student Research Committee, USERN Office, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Hojjatollah Nazari
- School of Biomedical Engineering, University of Technology Sydney, Sydney, NSW, Australia
| | - 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
| | - Fatemeh Heidari
- Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran
- Department of Anatomy, Faculty of Medicine, Qom University of Medical Sciences, Qom, Iran
| |
Collapse
|
48
|
Amini H, Namjoo AR, Narmi MT, Mardi N, Narimani S, Naturi O, Khosrowshahi ND, Rahbarghazi R, Saghebasl S, Hashemzadeh S, Nouri M. Exosome-bearing hydrogels and cardiac tissue regeneration. Biomater Res 2023; 27:99. [PMID: 37803483 PMCID: PMC10559618 DOI: 10.1186/s40824-023-00433-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 09/18/2023] [Indexed: 10/08/2023] Open
Abstract
BACKGROUND In recent years, cardiovascular disease in particular myocardial infarction (MI) has become the predominant cause of human disability and mortality in the clinical setting. The restricted capacity of adult cardiomyocytes to proliferate and restore the function of infarcted sites is a challenging issue after the occurrence of MI. The application of stem cells and byproducts such as exosomes (Exos) has paved the way for the alleviation of cardiac tissue injury along with conventional medications in clinics. However, the short lifespan and activation of alloreactive immune cells in response to Exos and stem cells are the main issues in patients with MI. Therefore, there is an urgent demand to develop therapeutic approaches with minimum invasion for the restoration of cardiac function. MAIN BODY Here, we focused on recent data associated with the application of Exo-loaded hydrogels in ischemic cardiac tissue. Whether and how the advances in tissue engineering modalities have increased the efficiency of whole-based and byproducts (Exos) therapies under ischemic conditions. The integration of nanotechnology and nanobiology for designing novel smart biomaterials with therapeutic outcomes was highlighted. CONCLUSION Hydrogels can provide suitable platforms for the transfer of Exos, small molecules, drugs, and other bioactive factors for direct injection into the damaged myocardium. Future studies should focus on the improvement of physicochemical properties of Exo-bearing hydrogel to translate for the standard treatment options.
Collapse
Affiliation(s)
- Hassan Amini
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of General and Vascular Surgery, Tabriz University of Medical Sciences, Tabriz, 51548/53431, Iran
| | - Atieh Rezaei Namjoo
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Maryam Taghavi Narmi
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Narges Mardi
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Samaneh Narimani
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ozra Naturi
- Department of Organic and Biochemistry, Faculty of Chemistry, University of Tabriz, Tabriz, Iran
| | - Nafiseh Didar Khosrowshahi
- Stem Cell and Tissue Engineering Research Laboratory, Sahand University of Technology, Tabriz, 51335-1996, Iran
| | - Reza Rahbarghazi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
- Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, 51548/53431, Iran.
| | - Solmaz Saghebasl
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
- Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, 51548/53431, Iran.
| | - Shahriar Hashemzadeh
- Department of General and Vascular Surgery, Tabriz University of Medical Sciences, Tabriz, 51548/53431, Iran.
| | - Mohammad Nouri
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
49
|
Vasella M, Arnke K, Dranseikiene D, Guzzi E, Melega F, Reid G, Klein HJ, Schweizer R, Tibbitt MW, Kim BS. Methacrylated Gelatin as a Scaffold for Mechanically Isolated Stromal Vascular Fraction for Cutaneous Wound Repair. Int J Mol Sci 2023; 24:13944. [PMID: 37762247 PMCID: PMC10530931 DOI: 10.3390/ijms241813944] [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: 08/03/2023] [Revised: 09/05/2023] [Accepted: 09/08/2023] [Indexed: 09/29/2023] Open
Abstract
Mechanically processed stromal vascular fraction (mSVF) is a highly interesting cell source for regenerative purposes, including wound healing, and a practical alternative to enzymatically isolated SVF. In the clinical context, SVF benefits from scaffolds that facilitate viability and other cellular properties. In the present work, the feasibility of methacrylated gelatin (GelMA), a stiffness-tunable, light-inducible hydrogel with high biocompatibility is investigated as a scaffold for SVF in an in vitro setting. Lipoaspirates from elective surgical procedures were collected and processed to mSVF and mixed with GelMA precursor solutions. Non-encapsulated mSVF served as a control. Viability was measured over 21 days. Secreted basic fibroblast growth factor (bFGF) levels were measured on days 1, 7 and 21 by ELISA. IHC was performed to detect VEGF-A, perilipin-2, and CD73 expression on days 7 and 21. The impact of GelMA-mSVF on human dermal fibroblasts was measured in a co-culture assay by the same viability assay. The viability of cultured GelMA-mSVF was significantly higher after 21 days (p < 0.01) when compared to mSVF alone. Also, GelMA-mSVF secreted stable levels of bFGF over 21 days. While VEGF-A was primarily expressed on day 21, perilipin-2 and CD73-positive cells were observed on days 7 and 21. Finally, GelMA-mSVF significantly improved fibroblast viability as compared with GelMA alone (p < 0.01). GelMA may be a promising scaffold for mSVF as it maintains cell viability and proliferation with the release of growth factors while facilitating adipogenic differentiation, stromal cell marker expression and fibroblast proliferation.
Collapse
Affiliation(s)
- Mauro Vasella
- Department of Plastic Surgery and Hand Surgery, University Hospital Zurich, 8091 Zurich, Switzerland; (M.V.); (G.R.)
| | - Kevin Arnke
- Center for Preclinical Development, University Hospital Zurich, 8091 Zurich, Switzerland;
| | - Dalia Dranseikiene
- Macromolecular Engineering Laboratory, Department of Mechanical and Process Engineering, ETH Zurich, 8092 Zurich, Switzerland; (D.D.); (E.G.); (M.W.T.)
| | - Elia Guzzi
- Macromolecular Engineering Laboratory, Department of Mechanical and Process Engineering, ETH Zurich, 8092 Zurich, Switzerland; (D.D.); (E.G.); (M.W.T.)
| | - Francesca Melega
- Institute of Pathology and Molecular Pathology, University Hospital Zurich, 8091 Zurich, Switzerland;
| | - Gregory Reid
- Department of Plastic Surgery and Hand Surgery, University Hospital Zurich, 8091 Zurich, Switzerland; (M.V.); (G.R.)
| | - Holger Jan Klein
- Department of Plastic Surgery and Hand Surgery, Cantonal Hospital Aarau, 5001 Aarau, Switzerland;
| | - Riccardo Schweizer
- Department of Plastic, Reconstructive and Aesthetic Surgery, Regional Hospital Lugano, 6900 Lugano, Switzerland;
| | - Mark W. Tibbitt
- Macromolecular Engineering Laboratory, Department of Mechanical and Process Engineering, ETH Zurich, 8092 Zurich, Switzerland; (D.D.); (E.G.); (M.W.T.)
| | - Bong-Sung Kim
- Department of Plastic Surgery and Hand Surgery, University Hospital Zurich, 8091 Zurich, Switzerland; (M.V.); (G.R.)
| |
Collapse
|