1
|
Tao Y, Liu T, Jing F, Tan X, Zhao X, Bernaerts KV, Jia R, Zhao J, Yin Y, Zhang T. Adipose-Derived Stem-Cell-Derived Exosomes Encapsulated Patch for Modulating Inflammation and Promoting Tissue Regeneration. ACS NANO 2025; 19:21271-21289. [PMID: 40470843 DOI: 10.1021/acsnano.4c16058] [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: 06/18/2025]
Abstract
Repairing tissue, especially in chronic wounds, is a major clinical challenge. Traditional treatments often lead to slow recovery and poor biocompatibility. Adipose-derived stem cell exosomes (ADSC-exo) offer a promising solution for tissue regeneration due to their anti-inflammatory and immunomodulatory effects. However, directly injecting stem cells results in a low survival and function loss. This study introduces a Janus G-Avs patch that promotes tissue repair by releasing ADSC-exo and prevents postoperative tissue adhesion. The patch's top layer, created via coaxial electrospinning, acts as an ADSC-exo delivery system with a core-shell structure by incorporating ADSC-exo and hyaluronic acid (HA) into methacrylate gelatin (GelMA) to prolong exosome release. The bottom layer consists of a 4arm-PLGA(4aPLGA)-Glu/PCL electrospun membrane, offering a lubricated antifouling surface that prevents protein adsorption and provides mechanical support. In rat models, the G-Avs patch has demonstrated its ability to enhance cell proliferation, promote angiogenesis, and encourage macrophage polarization toward the M2 phenotype, thereby mitigating inflammatory responses and promoting tissue repair. Transcriptomic analysis indicates that the G-Avs patch augments cellular energy metabolism by upregulating metabolic pathways and downregulating immune-inflammatory pathways, collectively supporting tissue repair. In summary, the Janus G-Avs patch has a strong clinical potential.
Collapse
Affiliation(s)
- Yinghua Tao
- State Key Laboratory of Digital Medicine Engineering, Southeast University, Nanjing 210096, China
- Institute of Medical Devices, Southeast University, Suzhou 215163, China
| | - Tao Liu
- State Key Laboratory of Digital Medicine Engineering, Southeast University, Nanjing 210096, China
- Institute of Medical Devices, Southeast University, Suzhou 215163, China
| | - Fengya Jing
- State Key Laboratory of Digital Medicine Engineering, Southeast University, Nanjing 210096, China
- Institute of Medical Devices, Southeast University, Suzhou 215163, China
| | - Xin Tan
- State Key Laboratory of Digital Medicine Engineering, Southeast University, Nanjing 210096, China
- Institute of Medical Devices, Southeast University, Suzhou 215163, China
| | - Xiaocong Zhao
- State Key Laboratory of Digital Medicine Engineering, Southeast University, Nanjing 210096, China
- Institute of Medical Devices, Southeast University, Suzhou 215163, China
| | - Katrien V Bernaerts
- Aachen-Maastricht Institute for Biobased Materials (AMIBM), Brightlands Chemelot Campus, Maastricht University, Urmonderbaan 22, 6167 RD Geleen, The Netherlands
| | - Ruipeng Jia
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing 210006, China
| | - Jun Zhao
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing 210006, China
| | - Yi Yin
- Department of General Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Tianzhu Zhang
- State Key Laboratory of Digital Medicine Engineering, Southeast University, Nanjing 210096, China
- Institute of Medical Devices, Southeast University, Suzhou 215163, China
| |
Collapse
|
2
|
Wu P, Zhang C, Wu C, Lang J, He L. Peimine Promotes Skin Wound Repair in Mice by Activating the Notch1 Signaling Pathway in Fibroblasts. Food Sci Nutr 2025; 13:e70406. [PMID: 40491981 PMCID: PMC12146499 DOI: 10.1002/fsn3.70406] [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: 09/24/2024] [Revised: 04/17/2025] [Accepted: 05/08/2025] [Indexed: 06/11/2025] Open
Abstract
This study aimed to investigate the effects of peimine on cutaneous wound healing efficiency and tissue regeneration in murine models, while exploring its regulatory mechanisms through the Notch1 signaling pathway. Full-thickness circular skin defects (8-mm diameter) were surgically created on the dorsal surface of C57BL/6 mice, with subsequent daily topical administration of peimine at 1 and 4 mg · kg-1 doses. Parallel in vitro experiments using NIH/3T3 fibroblasts were conducted with peimine treatments at 25 and 100 μmol · L-1 concentrations to assess neurogenic locus notch homolog protein 1 (Notch1) pathway activation. Key findings demonstrated that peimine treatment significantly enhanced both the rate of wound closure and granulation tissue formation. Histological analysis revealed increased epidermal thickness in peimine-treated groups compared to controls. The compound promoted extracellular matrix remodeling in the dermal layer, evidenced by elevated protein expression of collagen IIIα1 (Col3α1), proliferating cell nuclear antigen (PCNA), and the endothelial marker (CD31). Western blot analysis confirmed consistent upregulation of Notch1 pathway components in both in vivo wound tissues and in vitro fibroblast cultures, indicating that peimine accelerates wound repair through Notch1 signaling activation.
Collapse
Affiliation(s)
- Peng Wu
- Department of OrthopaedicsThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Cheng Zhang
- Department of OrthopaedicsThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Congcong Wu
- Department of OrthopaedicsThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Junzhe Lang
- Department of OrthopaedicsThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Lili He
- Department of DermatologyThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouChina
| |
Collapse
|
3
|
Liu C, Cheng C, Cheng K, Gao AS, Li Q, Atala A, Zhang Y. Precision exosome engineering for enhanced wound healing and scar revision. J Transl Med 2025; 23:578. [PMID: 40410904 PMCID: PMC12103044 DOI: 10.1186/s12967-025-06578-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2025] [Accepted: 05/05/2025] [Indexed: 05/25/2025] Open
Abstract
The dysfunction of wound-healing processes can result in chronic non-healing wounds and pathological scar formation. Current treatment options often fall short, necessitating innovative approaches. Exosomes, extracellular vesicles secreted by various cells, have emerged as promising therapeutic agents serving as an intercellular communication system. By engineering exosomes, their cargo and surface properties can be tailored to enhance therapeutic efficacy and specificity. Engineered exosomes (eExo) are emerging as a favorable tool for treating non-healing wounds and pathological scars. In this review, we delve into the underlying mechanisms of non-healing wounds and pathological scars, outline the current state of engineering strategies, and explore the clinical potential of eExo based on preclinical and clinical studies. In addition, we address the current challenges and future research directions, including standardization, safety and efficacy assessments, and potential immune responses. In conclusion, eExo hold great promise as a novel therapeutic approach for non-healing wounds and non-healing wounds and pathological scars. Further research and clinical trials are warranted to translate preclinical findings into effective clinical treatments.
Collapse
Affiliation(s)
- Chuanqi Liu
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Road, Shanghai, 200011, China
| | - Chen Cheng
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Road, Shanghai, 200011, China
| | - Kun Cheng
- Division of Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, Kansas City, MO, 64108-2718, USA
| | - Allen S Gao
- Department of Urologic Surgery, School of Medicine, University of California, Davis Sacramento, CA, 95817, USA
| | - Qingfeng Li
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Road, Shanghai, 200011, China.
| | - Anthony Atala
- Institute for Regenerative Medicine, Wake Forest University Health Sciences, Winston-Salem, NC, 27151, USA
| | - Yuanyuan Zhang
- Institute for Regenerative Medicine, Wake Forest University Health Sciences, Winston-Salem, NC, 27151, USA.
| |
Collapse
|
4
|
Thouvenot E, Charnay L, Burshtein N, Guigner J, Dec L, Loew D, Silva AK, Lindner A, Wilhelm C. High-Yield Bioproduction of Extracellular Vesicles from Stem Cell Spheroids via Millifluidic Vortex Transport. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2412498. [PMID: 39530646 PMCID: PMC12087746 DOI: 10.1002/adma.202412498] [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] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 09/23/2024] [Indexed: 11/16/2024]
Abstract
Extracellular vesicles (EVs) are emerging as novel therapeutics, particularly in cancer and degenerative diseases. Nevertheless, from both market and clinical viewpoints, high-yield production methods using minimal cell materials are still needed. Herein, a millifluidic cross-slot chip is proposed to induce high-yield release of biologically active EVs from less than three million cells. Depending on the flow rate, a single vortex forms in the outlet channels, exposing transported cellular material to high viscous stresses. Importantly, the chip accommodates producer cells within their physiological environment, such as human mesenchymal stem cells (hMSCs) spheroids, while facilitating their visualization and individual tracking within the vortex. This precise control of viscous stresses at the spheroid level allows for the release of up to 30000 EVs per cell at a Reynolds number of ≈400, without compromising cellular integrity. Additionally, it reveals a threshold initiating EV production, providing evidence for a stress-dependent mechanism governing vesicle secretion. EVs mass-produced at high Reynolds displayed pro-angiogenic and wound healing capabilities, as confirmed by proteomic and cytometric analysis of their cargo. These distinct molecular signatures of these EVs, compared to those derived from monolayers, underscore the critical roles of the production method and the 3D cellular environment in EV generation.
Collapse
Affiliation(s)
- Elliot Thouvenot
- Laboratoire Physique des Cellules et CancerPCCCNRS UMR168Institut CurieSorbonne Université, PSL Research UniversityParis75005France
| | - Laura Charnay
- Laboratoire Physique des Cellules et CancerPCCCNRS UMR168Institut CurieSorbonne Université, PSL Research UniversityParis75005France
| | - Noa Burshtein
- Laboratoire de Physique et Mécanique des Milieux HétérogènesPMMHCNRS UMR7636ESPCI ParisPSL Research UniversitySorbonne UniversitéUniversité Paris CitéParis75005France
| | - Jean‐Michel Guigner
- Institut de Minéralogiede Physique des Matériaux et de Cosmochimie (IMPMC)UMR CNRS 7590MNHNIRD UR 206Campus JussieuSorbonne UniversitéCase courrier 115, 4 Place Jussieu, 75252ParisCedex 05France
| | - Léonie Dec
- Institut CurieCurieCoreTech Mass Spectrometry ProteomicsPSL Research UniversityParisFrance
| | - Damarys Loew
- Institut CurieCurieCoreTech Mass Spectrometry ProteomicsPSL Research UniversityParisFrance
| | - Amanda K.A. Silva
- Laboratoire Matière et Systèmes ComplexesMSC, CNRS UMR7057, Université Paris CitéParis75006France
| | - Anke Lindner
- Laboratoire de Physique et Mécanique des Milieux HétérogènesPMMHCNRS UMR7636ESPCI ParisPSL Research UniversitySorbonne UniversitéUniversité Paris CitéParis75005France
| | - Claire Wilhelm
- Laboratoire Physique des Cellules et CancerPCCCNRS UMR168Institut CurieSorbonne Université, PSL Research UniversityParis75005France
| |
Collapse
|
5
|
Bai N, Li Y, Xu Q, Yang T. Revolutionizing tissue regeneration: the art of mimicking diverse collagen-based extracellular matrix hierarchies with decellularized tendon via bioskiving. Int J Biol Macromol 2025; 310:143532. [PMID: 40300687 DOI: 10.1016/j.ijbiomac.2025.143532] [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: 02/13/2025] [Revised: 04/16/2025] [Accepted: 04/25/2025] [Indexed: 05/01/2025]
Abstract
Precisely simulating the collagen-based extracellular matrix (ECM) hierarchy is essential for advancing tissue regeneration. Although decellularized ECM (dECM) demonstrates considerable potential in fostering tissue regeneration by maintaining the inherent hierarchy, its application remains restricted to homologous tissues due to hierarchical variability. The current challenge involves overcoming obstacles that impede the application of dECM across diverse tissues, with particular emphasis on re-engineering one type of dECM to mimic the complex hierarchies of other tissues. The decellularized tendon, distinguished by its aligned collagen fibers, has garnered substantial interest due to its favorable structural and mechanical properties. Recently, a novel technique termed bioskiving has been developed, which uniquely disassembles macroscale decellularized tendons into microscale collagen sheets via microtome sectioning, thereby preserving micro/nano architecture of fiber alignment without chemical denaturation. These sheets can be further rotationally stacked or rolled to engineer 2D or 3D constructs replicating diverse ECM hierarchies. Bioskiving effectively dismantles the persistent hindrance that traditionally impeded the application of dECM across various tissues. This review offers an in-depth analysis of the fabrication procedure and characterization of constructs produced through bioskiving. It specifically elucidates the applications of constructs across various domains in tissue engineering, demonstrating its potential in overcoming tissue-specific constraints of dECM.
Collapse
Affiliation(s)
- Na Bai
- Department of Prosthodontics, The Affiliated Hospital of Qingdao University, School of Stomatology of Qingdao University, Qingdao 266000, Shandong, China.; Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA..
| | - Yamin Li
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA.; Department of Pharmacology, State University of New York, Upstate Medical University, Syracuse, NY 13210, USA.
| | - Qiaobing Xu
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA..
| | - Tao Yang
- Department of Prosthodontics, Guanghua School of Stomatology, Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou 510055, Guangdong, China..
| |
Collapse
|
6
|
Rong J, Li YY, Wang X, Wang JN, Song M. Non-coding RNAs in adipose-derived stem cell exosomes: Mechanisms, therapeutic potential, and challenges in wound healing. World J Stem Cells 2025; 17:102917. [PMID: 40308889 PMCID: PMC12038460 DOI: 10.4252/wjsc.v17.i4.102917] [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: 11/05/2024] [Revised: 12/23/2024] [Accepted: 03/21/2025] [Indexed: 04/23/2025] Open
Abstract
The treatment of complex wounds presents a significant clinical challenge due to the limited availability of standardized therapeutic options. Adipose-derived stem cell exosomes (ADSC-Exos) are promising for their capabilities to enhance angiogenesis, mitigate oxidative stress, modulate inflammatory pathways, support skin cell regeneration, and promote epithelialization. These exosomes deliver non-coding RNAs, including microRNAs, long non-coding RNAs, and circular RNAs, which facilitate collagen remodeling, reduce scar formation, and expedite wound healing. This study reviews the mechanisms, therapeutic roles, and challenges of non-coding RNA-loaded ADSC-Exos in wound healing and identifies critical directions for future research. It aims to provide insights for researchers into the potential mechanisms and clinical applications of ADSC-Exos non-coding RNAs in wound healing.
Collapse
Affiliation(s)
- Jian Rong
- Department of Burns and Plastic Surgery, The 940 Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Lanzhou 730050, Gansu Province, China
- Department of Plateau Medicine, The 940 Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Lanzhou 730050, Gansu Province, China
| | - Yao-Yao Li
- Department of Burns and Plastic Surgery, The 940 Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Lanzhou 730050, Gansu Province, China
- Department of Plateau Medicine, The 940 Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Lanzhou 730050, Gansu Province, China
| | - Xin Wang
- Department of Burns and Plastic Surgery, The 940 Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Lanzhou 730050, Gansu Province, China
- Department of Plateau Medicine, The 940 Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Lanzhou 730050, Gansu Province, China
| | - Jia-Ning Wang
- Department of Burns and Plastic Surgery, The 940 Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Lanzhou 730050, Gansu Province, China
- Department of Plateau Medicine, The 940 Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Lanzhou 730050, Gansu Province, China
| | - Mei Song
- Department of Burns and Plastic Surgery, The 940 Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Lanzhou 730050, Gansu Province, China
- Department of Plateau Medicine, The 940 Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Lanzhou 730050, Gansu Province, China.
| |
Collapse
|
7
|
Ren S, Lv H, Chen S, Zhou J, Chen S, Chen J, Luo J, Guo Y, Wang H, Zhai J, Zhou Y. Photoresponsive Blood-Derived Protein Hydrogels Packed with Bioactive Carbon Dots Modulate Mitochondrial Homeostasis and Reprogram Metabolism for Chronic Wound Healing in Diabetes. ACS APPLIED MATERIALS & INTERFACES 2025; 17:20885-20900. [PMID: 40148098 DOI: 10.1021/acsami.5c00635] [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: 03/29/2025]
Abstract
Autologous platelet concentrates (APC) represent a class of personalized regenerative materials for vascularized tissue regeneration. However, shortcomings including poor controllability of gel formation, lack of reactive oxygen species (ROS) scavenging ability, and deficient anti-inflammatory capacity restrict the tissue healing outcomes of APC. This study proposes an APC-based synergistic platform (CurCDs@iPRF-MA) for the treatment of chronic wounds in diabetes. Such a platform is composed of injectable platelet-rich fibrin (iPRF), gelatin methacryloyl (GelMA), and a carbogenic nanodrug from curcumin (CurCDs) that is injectable before the light-induced gel formation process, greatly facilitating the clinical applications of APC. Significantly, CurCDs@iPRF-MA can modulate the mitochondrial homeostasis under inflammatory conditions, activate the oxidative phosphorylation (OXPHOS) program, and regulate the diabetic microenvironment through metabolic reprogramming to achieve macrophage phenotype regulation and ROS elimination, as well as promote vascularization by releasing autologous growth factors, dramatically improving the healing efficacy of the chronic wounds in diabetes. This study offers a practical and effective approach to developing spatiotemporally controllable and multifunctional APC-based hydrogels for highly effective tissue regeneration.
Collapse
Affiliation(s)
- Sicong Ren
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun 130021, Jilin, China
- School of Stomatology, Jilin University, Changchun 130021, Jilin, China
| | - Huixin Lv
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun 130021, Jilin, China
- School of Stomatology, Jilin University, Changchun 130021, Jilin, China
| | - Sheng Chen
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun 130021, Jilin, China
- School of Stomatology, Jilin University, Changchun 130021, Jilin, China
| | - Jing Zhou
- School of Stomatology, Jilin University, Changchun 130021, Jilin, China
| | - Siyu Chen
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun 130021, Jilin, China
- School of Stomatology, Jilin University, Changchun 130021, Jilin, China
| | - Jingxia Chen
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun 130021, Jilin, China
- School of Stomatology, Jilin University, Changchun 130021, Jilin, China
| | - Jiaxin Luo
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun 130021, Jilin, China
- School of Stomatology, Jilin University, Changchun 130021, Jilin, China
| | - Yuanxin Guo
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun 130021, Jilin, China
- School of Stomatology, Jilin University, Changchun 130021, Jilin, China
| | - Huan Wang
- State Key Laboratory of Rare Earth Resources Utilization and Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, P. R. China
| | - Jingjie Zhai
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun 130021, Jilin, China
- School of Stomatology, Jilin University, Changchun 130021, Jilin, China
| | - Yanmin Zhou
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun 130021, Jilin, China
- School of Stomatology, Jilin University, Changchun 130021, Jilin, China
| |
Collapse
|
8
|
Shi X, Wang Z, Bi Z, Liu J, Zhang P. Enhancing Immunomodulation and Osseointegration of Bone Implants via Thrombin-Activated Platelet-Rich Plasma Self-Assembly. Adv Healthc Mater 2025; 14:e2404295. [PMID: 39887661 DOI: 10.1002/adhm.202404295] [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: 11/01/2024] [Revised: 01/14/2025] [Indexed: 02/01/2025]
Abstract
Platelet-rich plasma (PRP) is characterized by elevated concentrations of growth factors that facilitate bone repair. Nonetheless, the effective integration of PRP with bone implants and the sustained release of its active constituents pose significant challenges. In this study, thrombin is grafted onto the surface of polyetheretherketone (PEEK) via an N,N'-Disuccinimidyl Carbonate (DSC) linker and the retained enzymatic activity of thrombin enables the controlled activation of PRP self-assembly, resulting in the formation of a functional bio-gel layer. The optimal thrombin concentration to be 100 U/ mL-1 is determined, at which point both the grafting amount and enzymatic activity of thrombin reaches their peak, with no further increases observed at higher concentrations. PRP solutions with varying platelet enrichment ratios are subsequently activated on the thrombin-grafted PEEK surface, yielding self-assembled bio-gels capable of sustained growth factor release for up to 16 days. The thrombin-activated PRP bio-gel on PEEK surface not only enhances in vitro cell adhesion, proliferation, osteogenic differentiation, vascularization and specific polarization of macrophages, but also effectively facilitates in vivo angiogenesis, immunomodulation and bone formation in a platelet dose-dependent manner. Consequently, the thrombin-activated PRP gel presents a promising strategy for the biological functionalization of PEEK implants in orthopedic applications.
Collapse
Affiliation(s)
- Xiaotong Shi
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
- Department of Orthopaedics, Capital Medical University Affiliated Beijing Friendship Hospital, Beijing, 100050, China
- Department of Orthopedic Surgery, The First Hospital of Jilin University, Changchun, 130021, China
| | - Zongliang Wang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
| | - Zhiguo Bi
- Department of Orthopedic Surgery, The First Hospital of Jilin University, Changchun, 130021, China
| | - Jianguo Liu
- Department of Orthopedic Surgery, The First Hospital of Jilin University, Changchun, 130021, China
| | - Peibiao Zhang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
| |
Collapse
|
9
|
Yoo D, Jung SY, Go D, Park JY, You DG, Jung WK, Li Y, Ding J, Park JH, Um W. Functionalized extracellular vesicles of mesenchymal stem cells for regenerative medicine. J Nanobiotechnology 2025; 23:219. [PMID: 40102934 PMCID: PMC11921732 DOI: 10.1186/s12951-025-03300-6] [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: 06/09/2024] [Accepted: 03/06/2025] [Indexed: 03/20/2025] Open
Abstract
Stem cell-derived extracellular vesicles (EVs) have emerged as a safe and potent alternative to regenerative medicine in recent decades. Furthermore, the adjustment of EV functions has been recently enabled by certain stem cell preconditioning methods, providing an exceptional opportunity to enhance the therapeutic potential or confer additional functions of stem cell-derived EVs. In this review, we discuss the recent progress of functionalized EVs, based on stem cell preconditioning, for treating various organ systems, such as the musculoskeletal system, nervous system, integumentary system, cardiovascular system, renal system, and respiratory system. Additionally, we summarize the expected outcomes of preconditioning methods for stem cells and their EVs. With recent progress, we suggest considerations and future directions for developing personalized medicine based on preconditioned stem cell-derived EVs.
Collapse
Affiliation(s)
- Donghyeon Yoo
- Department of Biotechnology, College of Fisheries Science, Pukyong National University, Busan, 48513, Republic of Korea
| | - Se Young Jung
- Department of Biotechnology, College of Fisheries Science, Pukyong National University, Busan, 48513, Republic of Korea
| | - Dabin Go
- Department of Biotechnology, College of Fisheries Science, Pukyong National University, Busan, 48513, Republic of Korea
| | - Ji Yeong Park
- Department of Biotechnology, College of Fisheries Science, Pukyong National University, Busan, 48513, Republic of Korea
| | - Dong Gil You
- Department of Chemical Engineering & Biotechnology, Tech University of Korea, Siheung, 15073, Republic of Korea
| | - Won-Kyo Jung
- Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan, 48513, Republic of Korea
- Major of Biomedical Engineering, Division of Smart Healthcare, College of Information Technology and Convergence and New-senior Healthcare Innovation Center (BK21 Plus), Pukyong National University, Busan, 48513, Republic of Korea
| | - Yuce Li
- College of Life Science and Health, Wuhan University of Science and Technology (WUST), Wuhan, 430065, China
| | - Jianxun Ding
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China
| | - Jae Hyung Park
- School of Chemical Engineering, College of Engineering, Sungkyunkwan University (SKKU), Suwon, 16419, Republic of Korea.
| | - Wooram Um
- Department of Biotechnology, College of Fisheries Science, Pukyong National University, Busan, 48513, Republic of Korea.
| |
Collapse
|
10
|
Luo L, Yang H, Huang J, Chen D, He Y, Lin J, Zeng H, Hua C, Lin Z, Wu M, Ma Y, Deng Q, Liu M, Li S. Airway basal stem cell-derived extracellular vesicles modulate proliferation, migration and collagen deposition of fibroblasts. Stem Cell Res Ther 2025; 16:140. [PMID: 40102996 PMCID: PMC11921531 DOI: 10.1186/s13287-025-04268-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Accepted: 03/06/2025] [Indexed: 03/20/2025] Open
Abstract
BACKGROUND Human bronchial epithelial cell-derived extracellular vesicles have demonstrated the ability to attenuate fibroblasts activation. However, the specific key effector cell populations mediating this inhibitory effect remain unidentified. Airway basal stem cells (BSCs), which serve as progenitor cells for bronchial epithelial cells, play a critical role in fibrotic remodeling processes and possess significant therapeutic potential. This study aimed to characterize BSC-derived extracellular vesicles (BSC-EVs) and investigate their regulatory influence on fibroblasts behavior. METHODS Airway BSCs were collected through bronchoscopic brushing and differential centrifugation. Fibroblasts were subsequently treated with BSC-EVs at various concentrations to evaluate their dose- and time-dependent effects in vitro. The proteomic composition of BSC-EVs was analyzed using four-dimensional data-independent acquisition quantitative mass spectrometry (4D-DIA). Moreover, a bleomycin-induced pulmonary fibrosis model was established to evaluate the safety and preliminary efficacy of BSC-EVs. RESULTS We successfully isolated and identified BSC-EVs, which expressed the nucleus-specific marker TP63, indicative of BSCs, but lacked the BSC marker KRT5. Our findings demonstrated that BSC-EVs enhanced fibroblasts proliferation and migration in a dose-dependent manner. Importantly, BSC-EVs significantly attenuated fibroblasts activation and promoted fibroblasts senescence. Utilizing 4D-DIA quantitative proteomics, we revealed that BSC-EVs modulate extracellular matrix remodeling processes and regulate the expression of key proteins, including collagen I/III and matrix metalloproteinases. Animal models utilizing intratracheal administration of BSC-EVs demonstrate efficient reduction of collagen deposition. CONCLUSION This study offers an extensive characterization of BSC-EVs, adhering to the guidelines set forth by MISEV2023. The findings underscore the significant therapeutic potential of BSC-EVs in the management of fibrotic diseases.
Collapse
Affiliation(s)
- Lisi Luo
- Guangzhou Institute of Respiratory Health, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
- Guangzhou National Laboratory, Guangzhou International Bio Island, No. 9 XingDaoHuanBei Road, Guangzhou, 510005, Guangdong Province, China
- Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Huijie Yang
- Guangzhou Institute of Respiratory Health, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
- Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Junfeng Huang
- Guangzhou Institute of Respiratory Health, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
- Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Difei Chen
- Guangzhou Institute of Respiratory Health, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
- Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Yushan He
- Guangzhou Institute of Respiratory Health, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
- Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Jinsheng Lin
- Guangzhou Institute of Respiratory Health, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
- Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Haikang Zeng
- Guangzhou Institute of Respiratory Health, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Chu Hua
- Guangzhou Medical University, Guangzhou, Guangdong, China
- Translatiaonl Research Centre of Regenrative Medicine and 3D Printing Technologies, Guangzhou Medical University, Guangzhou, China
| | - Zikai Lin
- Guangzhou Institute of Respiratory Health, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
- Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Minting Wu
- Guangzhou Institute of Respiratory Health, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Yuqin Ma
- Guangzhou Institute of Respiratory Health, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
- Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Qilin Deng
- Guangzhou Institute of Respiratory Health, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
- Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Ming Liu
- Guangzhou Institute of Respiratory Health, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China.
| | - Shiyue Li
- Guangzhou Institute of Respiratory Health, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
11
|
Liu J, Li Y, Zhang Y, Zhao Z, Liu B. Engineered stromal vascular fraction for tissue regeneration. Front Pharmacol 2025; 16:1510508. [PMID: 40183080 PMCID: PMC11966044 DOI: 10.3389/fphar.2025.1510508] [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: 10/13/2024] [Accepted: 02/19/2025] [Indexed: 04/05/2025] Open
Abstract
The treatment of various tissue injuries presents significant challenges, particularly in the reconstruction of large and severe tissue defects, with conventional clinical methods often yielding suboptimal results. However, advances in engineering materials have introduced new possibilities for tissue repair. Bioactive components are commonly integrated with synthetic materials to enhance tissue reconstruction. Stromal vascular fraction (SVF), an adipose-derived cell cluster, has shown considerable potential in tissue regeneration due to its simple and efficient way of obtaining and its richness in growth factors. Therefore, this review illustrated the preparation, characterization, mechanism of action, and applications of engineered SVF in various tissue repair processes, to provide some references for the option of better methods for tissue defect reconstruction.
Collapse
Affiliation(s)
- Jianfeng Liu
- Department of Hand and Foot Surgery, Orthopedics Center, The First Hospital of Jilin University, Changchun, China
- Engineering Laboratory of Tissue Engineering Biomaterials of Jilin Province, Changchun, China
| | - Yiwei Li
- Department of Hand and Foot Surgery, Orthopedics Center, The First Hospital of Jilin University, Changchun, China
- Engineering Laboratory of Tissue Engineering Biomaterials of Jilin Province, Changchun, China
| | - Yanan Zhang
- Department of Hand and Foot Surgery, Orthopedics Center, The First Hospital of Jilin University, Changchun, China
- Engineering Laboratory of Tissue Engineering Biomaterials of Jilin Province, Changchun, China
| | - Zhiwei Zhao
- Department of Hand and Foot Surgery, Orthopedics Center, The First Hospital of Jilin University, Changchun, China
- Engineering Laboratory of Tissue Engineering Biomaterials of Jilin Province, Changchun, China
| | - Bin Liu
- Department of Hand and Foot Surgery, Orthopedics Center, The First Hospital of Jilin University, Changchun, China
- Engineering Laboratory of Tissue Engineering Biomaterials of Jilin Province, Changchun, China
| |
Collapse
|
12
|
Xu M, Zhang M, Wu J, Wang J, Wu H, Xu X. Esculin promotes skin wound healing in mice and regulates the Wnt/β-catenin signaling pathway. Cytojournal 2025; 22:32. [PMID: 40260065 PMCID: PMC12010908 DOI: 10.25259/cytojournal_184_2024] [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: 09/10/2024] [Accepted: 01/25/2025] [Indexed: 04/23/2025] Open
Abstract
Objective Previous studies reported that esculin could protect against renal ischemia-reperfusion injury and liver injury, but its mechanism of action in skin wound healing is unclear. The Wnt/β-catenin signaling pathway plays a positive role in the wound healing process. This study aimed to investigate the effects of esculin on the rate and quality of skin wound healing in mice and explore its regulatory role in the Wnt/b-catenin signaling pathway. Material and Methods Circular full-thickness skin wounds with a diameter of 8 mm were created on the backs of C57BL/6 mice, which were administered with 20 and 40 mg•kg-1 esculin through gastric lavage. Wound healing was monitored, and samples collected on day 14 were analyzed through hematoxylin-eosin and Masson staining to assess granulation tissue formation and collagen deposition. Immunohistochemistry, immunofluorescence, and Western blot evaluated markers of collagen synthesis, proliferation, angiogenesis, and proteins in the Wnt/b-catenin signaling pathway. National institutes of health/3T3 cells treated with esculin (50 and 200 μM) were analyzed for proliferating cell nuclear antigen (PCNA) expression to assess proliferative activity. Results Compared with the model group, the esculin-treated groups exhibited significantly enhanced wound healing (P < 0.05), increased skin epithelial thickness (P < 0.01), and promoted extracellular matrix formation in mice. In addition, esculin significantly raised type I collagen alpha-1 chain and type III collagen alpha-1 chain protein levels (P < 0.05), boosted the expression of the cell proliferation marker PCNA and the vascular marker cluster of differentiation 31 in the dermis (P < 0.05), and upregulated proteins related to the Wnt/b-catenin signaling pathway and increased glycogen synthase kinase 3 beta phosphorylation in skin wound and NIH/3T3 cells (P < 0.05). Conclusion Esculin could upregulate and activate the Wnt/b-catenin signaling pathway to promote wound healing.
Collapse
Affiliation(s)
- Mian Xu
- Department of Dermatology, Wenzhou Central Hospital, Wenzhou, China
| | - Mengsi Zhang
- Department of Obstetrics and Gynecology, Wenzhou Central Hospital, Wenzhou, China
| | - Jingjing Wu
- Department of Dermatology, Wenzhou Central Hospital, Wenzhou, China
| | - Jinmeng Wang
- Department of Dermatology, Wenzhou Central Hospital, Wenzhou, China
| | - Huaze Wu
- Department of Dermatology, Wenzhou Central Hospital, Wenzhou, China
| | - Xianting Xu
- Department of Dermatology, Wenzhou Central Hospital, Wenzhou, China
| |
Collapse
|
13
|
Yang Y, Deng C, Aldali F, Huang Y, Luo H, Liu Y, Huang D, Cao X, Zhou Q, Xu J, Li Y, Chen H. Therapeutic Approaches and Potential Mechanisms of Small Extracellular Vesicles in Treating Vascular Dementia. Cells 2025; 14:409. [PMID: 40136659 PMCID: PMC11941715 DOI: 10.3390/cells14060409] [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: 01/12/2025] [Revised: 02/26/2025] [Accepted: 03/10/2025] [Indexed: 03/27/2025] Open
Abstract
Small extracellular vesicles (sEVs), including exosomes as a subtype, with a diameter typically less than 200 nm and originating from the endosomal system, are capable of transporting a diverse array of bioactive molecules, including proteins, nucleic acids, and lipids, thereby facilitating intercellular communication and modulating cellular functions. Vascular dementia (VaD) represents a form of cognitive impairment attributed to cerebrovascular disease, characterized by a complex and multifaceted pathophysiological mechanism. Currently, the therapeutic approach to VaD predominantly emphasizes symptom management, as no specific pharmacological treatment exists to cure the condition. Recent investigations have illuminated the significant role of sEVs in the pathogenesis of vascular dementia. This review seeks to provide a comprehensive analysis of the characteristics and functions of sEVs, with a particular focus on their involvement in vascular dementia and its underlying mechanisms. The objective is to advance the understanding of the interplays between sEVs and vascular dementia, thereby offering novel insights for future research and therapeutic strategies.
Collapse
Affiliation(s)
- Yujie Yang
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Y.Y.); (C.D.); (F.A.); (Y.H.); (H.L.); (Y.L.); (D.H.); (X.C.); (Q.Z.); (J.X.); (Y.L.)
| | - Chunchu Deng
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Y.Y.); (C.D.); (F.A.); (Y.H.); (H.L.); (Y.L.); (D.H.); (X.C.); (Q.Z.); (J.X.); (Y.L.)
| | - Fatima Aldali
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Y.Y.); (C.D.); (F.A.); (Y.H.); (H.L.); (Y.L.); (D.H.); (X.C.); (Q.Z.); (J.X.); (Y.L.)
| | - Yunjie Huang
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Y.Y.); (C.D.); (F.A.); (Y.H.); (H.L.); (Y.L.); (D.H.); (X.C.); (Q.Z.); (J.X.); (Y.L.)
| | - Hongmei Luo
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Y.Y.); (C.D.); (F.A.); (Y.H.); (H.L.); (Y.L.); (D.H.); (X.C.); (Q.Z.); (J.X.); (Y.L.)
| | - Yizhou Liu
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Y.Y.); (C.D.); (F.A.); (Y.H.); (H.L.); (Y.L.); (D.H.); (X.C.); (Q.Z.); (J.X.); (Y.L.)
| | - Danxia Huang
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Y.Y.); (C.D.); (F.A.); (Y.H.); (H.L.); (Y.L.); (D.H.); (X.C.); (Q.Z.); (J.X.); (Y.L.)
| | - Xiaojian Cao
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Y.Y.); (C.D.); (F.A.); (Y.H.); (H.L.); (Y.L.); (D.H.); (X.C.); (Q.Z.); (J.X.); (Y.L.)
| | - Qiuzhi Zhou
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Y.Y.); (C.D.); (F.A.); (Y.H.); (H.L.); (Y.L.); (D.H.); (X.C.); (Q.Z.); (J.X.); (Y.L.)
| | - Jia Xu
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Y.Y.); (C.D.); (F.A.); (Y.H.); (H.L.); (Y.L.); (D.H.); (X.C.); (Q.Z.); (J.X.); (Y.L.)
- Stem Cell Research Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yajie Li
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Y.Y.); (C.D.); (F.A.); (Y.H.); (H.L.); (Y.L.); (D.H.); (X.C.); (Q.Z.); (J.X.); (Y.L.)
| | - Hong Chen
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Y.Y.); (C.D.); (F.A.); (Y.H.); (H.L.); (Y.L.); (D.H.); (X.C.); (Q.Z.); (J.X.); (Y.L.)
- Stem Cell Research Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan 430030, China
| |
Collapse
|
14
|
Youssef E, Palmer D, Fletcher B, Vaughn R. Exosomes in Precision Oncology and Beyond: From Bench to Bedside in Diagnostics and Therapeutics. Cancers (Basel) 2025; 17:940. [PMID: 40149276 PMCID: PMC11940788 DOI: 10.3390/cancers17060940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 02/28/2025] [Accepted: 03/07/2025] [Indexed: 03/29/2025] Open
Abstract
Exosomes have emerged as pivotal players in precision oncology, offering innovative solutions to longstanding challenges such as metastasis, therapeutic resistance, and immune evasion. These nanoscale extracellular vesicles facilitate intercellular communication by transferring bioactive molecules that mirror the biological state of their parent cells, positioning them as transformative tools for cancer diagnostics and therapeutics. Recent advancements in exosome engineering, artificial intelligence (AI)-driven analytics, and isolation technologies are breaking barriers in scalability, reproducibility, and clinical application. Bioengineered exosomes are being leveraged for CRISPR-Cas9 delivery, while AI models are enhancing biomarker discovery and liquid biopsy accuracy. Despite these advancements, key obstacles such as heterogeneity in exosome populations and the lack of standardized isolation protocols persist. This review synthesizes pioneering research on exosome biology, molecular engineering, and clinical translation, emphasizing their dual roles as both mediators of tumor progression and tools for intervention. It also explores emerging areas, including microbiome-exosome interactions and the integration of machine learning in exosome-based precision medicine. By bridging innovation with translational strategies, this work charts a forward-looking path for integrating exosomes into next-generation cancer care, setting it apart as a comprehensive guide to overcoming clinical and technological hurdles in this rapidly evolving field.
Collapse
|
15
|
An W, Zhang W, Qi J, Xu W, Long Y, Qin H, Yao K. Mesenchymal stem cells and mesenchymal stem cell-derived exosomes: a promising strategy for treating retinal degenerative diseases. Mol Med 2025; 31:75. [PMID: 39984849 PMCID: PMC11846226 DOI: 10.1186/s10020-025-01120-w] [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: 12/30/2024] [Accepted: 02/07/2025] [Indexed: 02/23/2025] Open
Abstract
Mesenchymal stem cells (MSCs) have emerged as a promising therapeutic strategy in regenerative medicine, demonstrating significant potential for clinical applications. Evidence suggests that MSCs not only exhibit multipotent differentiation potential but also exert critical therapeutic effects in retinal degenerative diseases via robust paracrine mechanisms. MSCs protect retinal cells from degenerative damage by modulating inflammation, inhibiting apoptosis, alleviating oxidative stress, and suppressing cell death pathways. Furthermore, MSCs contribute to retinal structural and functional stability by facilitating vascular remodeling and donating mitochondria to retinal cells. Of particular interest, MSC-derived exosomes have gained widespread attention as a compelling cell-free therapy. Owing to their potent anti-inflammatory, anti-apoptotic, and vascular-stabilizing properties, exosomes show significant promise for the treatment of retinal degenerative diseases.
Collapse
Affiliation(s)
- Wenjing An
- Institute of Visual Neuroscience and Stem Cell Engineering, Wuhan University of Science and Technology, Wuhan, 430065, China
- College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan, 430065, China
| | - Wenliang Zhang
- Institute of Visual Neuroscience and Stem Cell Engineering, Wuhan University of Science and Technology, Wuhan, 430065, China
- College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan, 430065, China
| | - Jia Qi
- Institute of Visual Neuroscience and Stem Cell Engineering, Wuhan University of Science and Technology, Wuhan, 430065, China
- College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan, 430065, China
| | - Weihui Xu
- Institute of Visual Neuroscience and Stem Cell Engineering, Wuhan University of Science and Technology, Wuhan, 430065, China
- College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan, 430065, China
| | - Yushan Long
- Institute of Visual Neuroscience and Stem Cell Engineering, Wuhan University of Science and Technology, Wuhan, 430065, China
- College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan, 430065, China
| | - Huan Qin
- Institute of Visual Neuroscience and Stem Cell Engineering, Wuhan University of Science and Technology, Wuhan, 430065, China.
- College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan, 430065, China.
| | - Kai Yao
- Institute of Visual Neuroscience and Stem Cell Engineering, Wuhan University of Science and Technology, Wuhan, 430065, China.
- College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan, 430065, China.
| |
Collapse
|
16
|
Hong X, Cai L, Li L, Zheng D, Lin J, Liang Z, Fu W, Liang D, Zeng T, Sun K, Wang W, Chen S, Ren M, Yan L. Keratinocyte-derived small extracellular vesicles delay diabetic wound healing by triggering fibroblasts autophagy. Arch Physiol Biochem 2025; 131:11-23. [PMID: 38828847 DOI: 10.1080/13813455.2024.2358020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Accepted: 05/16/2024] [Indexed: 06/05/2024]
Abstract
Keratinocyte and fibroblast dysfunctions contribute to delayed healing of diabetic wounds. Small extracellular vesicles (sEV) are key mediators of intercellular communication and are involved in the pathogenesis of several diseases. Recent findings suggest that sEV derived from high-glucose-treated keratinocyte (HaCaT-HG-sEV) can transport LINC01435 to inhibit tube formation and migration of HUVECs, thereby delaying wound healing. This study aimed to elucidate sEV-related communication mechanisms between keratinocytes and fibroblasts during diabetic wound healing. HaCaT-HG-sEV treatment and LINC01435 overexpression significantly decreased fibroblast collagen level and migration ability but significantly increased fibroblast autophagy. However, treatment with an autophagy inhibitor suppressed LINC01435 overexpression-induced decrease in collagen levels in fibroblasts. In diabetic mice, HaCaT-HG-sEV treatment decreased collagen levels and increased the expression of the autophagy-related proteins Beclin-1 and LC3 at the wound site, thereby delaying wound healing. Conclusively, LINC01435 in keratinocyte-derived sEV activates fibroblast autophagy and reduces fibroblast collagen synthesis, leading to impaired diabetic wound healing.
Collapse
Affiliation(s)
- Xiaosi Hong
- Department of Endocrinology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Leiqin Cai
- Department of Endocrinology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Lanlan Li
- Department of Endocrinology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Dinghao Zheng
- Department of Endocrinology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Jianghong Lin
- Department of Endocrinology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Zhuoxian Liang
- Department of Endocrinology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Wan Fu
- Department of Endocrinology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Diefei Liang
- Department of Endocrinology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Tingting Zeng
- Department of Endocrinology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Kan Sun
- Department of Endocrinology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Wei Wang
- Department of Endocrinology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Department of Endocrinology, Shenshan Medical center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Sifan Chen
- Department of Endocrinology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan, China
| | - Meng Ren
- Department of Endocrinology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Li Yan
- Department of Endocrinology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
17
|
Lin YH, Chen Y, Liu EW, Chen MC, Yu MH, Chen CY, Ho CC, Hsu-Jiang TY, Lee JJ, Cho DY, Shie MY. Immunomodulation effects of collagen hydrogel encapsulating extracellular vesicles derived from calcium silicate stimulated-adipose mesenchymal stem cells for diabetic healing. J Nanobiotechnology 2025; 23:45. [PMID: 39865263 PMCID: PMC11770968 DOI: 10.1186/s12951-025-03097-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Accepted: 01/05/2025] [Indexed: 01/28/2025] Open
Abstract
Diabetic wounds are characterized by chronic inflammation, reduced angiogenesis, and insufficient collagen deposition, leading to impaired healing. Extracellular vesicles (EVs) derived from adipose-derived mesenchymal stem cells (ADSC) offer a promising cell-free therapeutic strategy, yet their efficacy and immunomodulation can be enhanced through bioactivation. In this study, we developed calcium silicate (CS)-stimulated ADSC-derived EVs (CSEV) incorporated into collagen hydrogels to create a sustained-release system for promoting diabetic wound healing. CSEV exhibited enhanced protein content, surface marker expression, and bioactive cargo enriched with pro-angiogenic and anti-inflammatory factors. In vitro, CSEV-loaded collagen significantly reduced reactive oxygen species production, promoted cell proliferation and migration compared to standard EV-loaded collagen. Cytokine profiling revealed the upregulation of anti-inflammatory cytokines and extracellular matrix components, highlighting their immunomodulatory and regenerative potential. In vivo, histological evaluation of diabetic rabbit models treated with CSEV-loaded collagen revealed superior reepithelialization and organized collagen deposition, indicating accelerated wound closure. These findings underscore the potential of CSEV-loaded collagen hydrogels as an innovative and effective therapeutic platform for enhancing diabetic wound healing by simultaneously addressing inflammation and tissue regeneration.
Collapse
Affiliation(s)
- Yen-Hong Lin
- Department of Biomedical Engineering, China Medical University, Taichung, 406040, Taiwan
- Research & Development Center for x-Dimensional Extracellular Vesicles, Department of Medical Research, China Medical University Hospital, Taichung, 404327, Taiwan
| | - Yeh Chen
- Department of Food Science and Biotechnology, National Chung Hsing University, Taichung, 402202, Taiwan
| | - En-Wei Liu
- Department of Plastic and Reconstructive Surgery, China Medical University Hospital, Taichung, 404327, Taiwan
| | - Mei-Chih Chen
- Translational Cell Therapy Center, Department of Medical Research, China Medical University Hospital, Taichung, 404327, Taiwan
- Department and Development Department, Shine Out Bio Technology Co., Ltd, Taichung, 407608, Taiwan
| | - Min-Hua Yu
- Institute of Translational Medicine and New Drug Development, China Medical University, Taichung, 406040, Taiwan
| | - Cheng-Yu Chen
- Research & Development Center for x-Dimensional Extracellular Vesicles, Department of Medical Research, China Medical University Hospital, Taichung, 404327, Taiwan
| | - Chia-Che Ho
- Department of Bioinformatics and Medical Engineering, Asia University, Taichung, 413305, Taiwan
- High Performance Materials Institute for x-Dimensional Printing, Asia University, Taichung, 413305, Taiwan
| | - Tai-Yi Hsu-Jiang
- School of Medicine, China Medical University, Taichung, 406040, Taiwan
| | - Jian-Jr Lee
- Department of Plastic and Reconstructive Surgery, China Medical University Hospital, Taichung, 404327, Taiwan.
- School of Medicine, China Medical University, Taichung, 406040, Taiwan.
| | - Der-Yang Cho
- Research & Development Center for x-Dimensional Extracellular Vesicles, Department of Medical Research, China Medical University Hospital, Taichung, 404327, Taiwan.
- Translational Cell Therapy Center, Department of Medical Research, China Medical University Hospital, Taichung, 404327, Taiwan.
- Department of Neurosurgery, China Medical University Hospital, Taichung, 404327, Taiwan.
| | - Ming-You Shie
- Department of Biomedical Engineering, China Medical University, Taichung, 406040, Taiwan.
- Research & Development Center for x-Dimensional Extracellular Vesicles, Department of Medical Research, China Medical University Hospital, Taichung, 404327, Taiwan.
- Department of Bioinformatics and Medical Engineering, Asia University, Taichung, 413305, Taiwan.
| |
Collapse
|
18
|
Jiang T, Liu Q, Xu EC, He SY, Liu HY, Tian C, Zhang LF, Yang ZL. Fibroblasts/three-dimensional scaffolds complexes promote wound healing in rats with skin defects. Tissue Barriers 2025; 13:2334544. [PMID: 38544287 PMCID: PMC11875492 DOI: 10.1080/21688370.2024.2334544] [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: 10/19/2023] [Revised: 03/11/2024] [Accepted: 03/20/2024] [Indexed: 03/03/2025] Open
Abstract
We aim to construct a three-dimensional nano-skin scaffold material in vitro and study its promoting effect on wound healing in vivo. In this study, hybrid constructs of three-dimensional (3D) scaffolds were successfully fabricated by combination of type I collagen (COL-1) and polylactic-glycolic acid (PLGA). Fibroblasts and human umbilical cord mesenchymal stem cells (hUCMSCs) were used to implanted into 3D scaffolds and constructed into SD skin scaffolds in vitro. Finally, the fibroblasts/scaffolds complexes were inoculated on the surface of rat wound skin to study the promoting effect of the complex on wound healing. In our study, we successfully built a 3D scaffold, which had a certain porosity. Meanwhile, the content of COL-1 in the cell supernatant of fibroblast/scaffold complexes was increased. Furthermore, the expression of F-actin, CD105, integrin β, VEGF, and COL-1 was up-regulated in hUCMSC/scaffold complexes compared with the control group. In vivo, fibroblast/scaffold complexes promoted wound healing in rats. Our data suggested that the collagen Ⅳ and vimentin were elevated and collagen fibers were neatly arranged in the fibroblast/scaffold complex group was significantly higher than that in the scaffold group. Taken together, fibroblast/scaffold complexes were expected to be novel materials for treating skin defects.
Collapse
Affiliation(s)
- Ting Jiang
- Department of Burn and Plastic Surgery, Nanchong Central Hospital, The Second Clinical College of North Sichuan Medical College, Nanchong, China
| | - Qiang Liu
- Department of Burn and Plastic Surgery, Nanchong Central Hospital, The Second Clinical College of North Sichuan Medical College, Nanchong, China
| | - Er-Chang Xu
- Department of Burn and Plastic Surgery, Nanchong Central Hospital, The Second Clinical College of North Sichuan Medical College, Nanchong, China
| | - Si-Yu He
- Department of Burn and Plastic Surgery, Nanchong Central Hospital, The Second Clinical College of North Sichuan Medical College, Nanchong, China
| | - Hong-Yan Liu
- Department of Burn and Plastic Surgery, Nanchong Central Hospital, The Second Clinical College of North Sichuan Medical College, Nanchong, China
| | - Chao Tian
- Department of Burn and Plastic Surgery, Nanchong Central Hospital, The Second Clinical College of North Sichuan Medical College, Nanchong, China
| | - Lan-Fang Zhang
- Department of Burn and Plastic Surgery, Nanchong Central Hospital, The Second Clinical College of North Sichuan Medical College, Nanchong, China
| | - Ze-Long Yang
- Department of Orthopaedic Surgery, Nanchong Central Hospital, The Second Clinical College of North Sichuan Medical College, Nanchong, China
| |
Collapse
|
19
|
Chen Y, Wang Q, Ning F, Du C, Chen M, Feng C, Dong CM. Dynamic Hyaluronic Acid Hydrogels for Comprehensively Regulating Inflammation, Angiogenesis, and Metabolism to Effectively Proheal Diabetic Wounds. ACS APPLIED MATERIALS & INTERFACES 2024; 16:70256-70273. [PMID: 39668760 DOI: 10.1021/acsami.4c15674] [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: 12/14/2024]
Abstract
Despite the great progress of various multifunctional wound dressings, it is challenging to simultaneously achieve complete healing and functional remodeling for diabetic foot ulcers and refractory chronic wounds. Aiming to comprehensively regulate chronic inflammation, angiogenesis, and metabolism processes, herein, a novel kind of dynamic hyaluronic acid (HA) hydrogel was designed by combining boronate and coordination chemistry. Besides having injectability, self-healing, and detachment properties, dynamic HA hydrogels presented diabetic wound-responsive degradation and controllable H2S release. They could efficiently polarize M1-to-M2 polarization and regulate inflammatory cytokine secretion and multiple inflammation-related mRNA expressions through cooperative actions of reactive oxygen species elimination + H2S release + Zn2+ regulation, thus driving chronic inflammation into the proliferation and remodeling stages. Moreover, the screened lead hydrogel HTZS could regulate angiogenesis-related signaling pathways and metabolism processes to promote neovascularization and mature vessel formation, re-epithelization, high-level collagen-I deposition, and dense hair follicle regeneration, achieving complete healing and functional remodeling in diabetic wounds. Importantly, this work opens a new avenue to design dynamic biopolymer hydrogels for high-performance wound dressing and decipher the key role of multiple orchestrated regulations of inflammation-angiogenesis-metabolism on complete healing and functional remodeling in chronic and diabetic wounds.
Collapse
Affiliation(s)
- Yanzheng Chen
- School of Chemistry and Chemical Engineering, Shanghai Key Laboratory of Electrical Insulation and Thermal Aging, Shanghai Jiao Tong University, Shanghai 200240, P. R. China
| | - Qing Wang
- Department of Stomatology, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai 200080, P. R. China
| | - Fangrui Ning
- School of Chemistry and Chemical Engineering, Shanghai Key Laboratory of Electrical Insulation and Thermal Aging, Shanghai Jiao Tong University, Shanghai 200240, P. R. China
| | - Chang Du
- Clinical Cancer Institute, Center for Translational Medicine, Naval Military Medical University, Shanghai 200433, P. R. China
| | - Mingsheng Chen
- Shanghai Public Health Clinical Center, Fudan University, 2901 Caolang Road, Shanghai 201508, P. R. China
| | - Chuanliang Feng
- School of Materials Science and Engineering, State Key Laboratory of Metal Matrix Composites, Shanghai Jiao Tong University, Shanghai 200240, P. R. China
| | - Chang-Ming Dong
- School of Chemistry and Chemical Engineering, Shanghai Key Laboratory of Electrical Insulation and Thermal Aging, Shanghai Jiao Tong University, Shanghai 200240, P. R. China
| |
Collapse
|
20
|
Ding Y, Song M, Huang R, Chen W. Adipose-mesenchymal stem cell-derived extracellular vesicles enhance angiogenesis and skin wound healing via bFGF-mediated VEGF expression. Cell Tissue Bank 2024; 26:2. [PMID: 39625539 DOI: 10.1007/s10561-024-10150-3] [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: 07/23/2024] [Accepted: 11/04/2024] [Indexed: 02/22/2025]
Abstract
This study aimed to investigate whether extracellular vesicles (EVs) derived from adipose-derived mesenchymal stem cells (ASCs) promote skin wound healing by delivering basic fibroblast growth factor (bFGF) to enhance vascular endothelial growth factor (VEGF) expression. ASCs were isolated and transfected with either a bFGF knockdown lentivirus (Lv-sh-bFGF) or a control lentivirus (Lv-sh-NC). EVs were extracted from ASCs cultures and characterized by transmission electron microscopy, nanoparticle tracking analysis, and Western blotting for surface markers. EVs were extracted from the conditioned mediums of ASCs and subjected to different treatments. These EVs or control treatments were injected at the wound edges. Wound healing was assessed using histological techniques, including H&E and Masson's trichrome staining to evaluate tissue regeneration, collagen organization, and immunohistochemistry for CD31 to quantify microvessel density. Protein expression of bFGF and VEGF was measured by Western blotting. ASC-derived EVs significantly promoted angiogenesis and improved skin wound healing. EVs encapsulating bFGF enhanced VEGF expression in the wound tissue, while knockdown of bFGF reduced both bFGF and VEGF expression, leading to delayed wound healing. Further knockdown of VEGF partially reversed the pro-angiogenic and wound-healing effects of bFGF-encapsulated EVs. This study demonstrates that ASC-derived EVs promoted skin wound repair by enhancing angiogenesis and accelerating tissue regeneration through the bFGF/VEGF axis. These findings highlight the therapeutic potential of ASCs-derived EVs in improving skin wound healing.
Collapse
Affiliation(s)
- Yonghu Ding
- Department of Orthopedics, The Third People's Hospital Health Care Group of Cixi, 51-139 Zhouxi Road, Zhouxiang Town, Cixi City, Ningbo, 315000, China
| | - Mengsheng Song
- Department of Orthopedics, The Third People's Hospital Health Care Group of Cixi, 51-139 Zhouxi Road, Zhouxiang Town, Cixi City, Ningbo, 315000, China
| | - Rong Huang
- Department of Orthopedics, The Third People's Hospital Health Care Group of Cixi, 51-139 Zhouxi Road, Zhouxiang Town, Cixi City, Ningbo, 315000, China
| | - Weiting Chen
- Department of Orthopedics, The Third People's Hospital Health Care Group of Cixi, 51-139 Zhouxi Road, Zhouxiang Town, Cixi City, Ningbo, 315000, China.
| |
Collapse
|
21
|
Li Y, Dong L, Chen Y, Cai W, Yang G, Wang Y. Epithelial differentiation of gingival mesenchymal stem cells enhances re-epithelialization for full-thickness cutaneous wound healing. Stem Cell Res Ther 2024; 15:455. [PMID: 39609719 PMCID: PMC11605919 DOI: 10.1186/s13287-024-04081-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 11/25/2024] [Indexed: 11/30/2024] Open
Abstract
BACKGROUND Increasing evidence suggests that mesenchymal stem cells (MSCs) repair traumatized tissues primarily through paracrine secretion and differentiation into specific cell types. However, the role of epithelial differentiation of MSCs in cutaneous wound healing is unclear. This study aimed to investigate the epithelial differentiation potential of gingival tissue-derived MSCs (GMSCs) in epithelial cell growth medium and the mechanisms underlying their differentiation into an epithelial-like cell phenotype. METHODS We used scanning electron microscopy to examine GMSCs for epithelial differentiation. Quantitative real-time polymerase chain reaction and Western blotting were respectively used to measure genes and proteins related to epithelial differentiation. Immunofluorescence was used to examine subcellular localization of KLF4, KRT19, and β-catenin proteins. Transcriptome sequencing was used to enrich the mechanisms underlying epithelial differentiation in GMSCs. An MSAB inhibitor was used to validate the Wnt signaling pathway further. The wound healing rate and re-epithelialization were assessed through macroscopical observation and hematoxylin and eosin staining. RESULTS GMSCs cultured in epithelial cell growth medium from days 3 to 15 exhibited decreased expression of mesenchymal-epithelial transition and stemness-related proteins (N-cadherin, Vimentin, KLF4, and SOX2), increased expression of epithelial-related proteins (KRT12, KRT15, KRT19, and E-cadherin), and exhibited epithelial-like morphology. Mechanistically, high-throughput sequencing revealed that the Wnt and TGF-beta signaling pathways were inhibited during epithelial differentiation of GMSCs (Epi-GMSCs). MSAB-induced Wnt signaling pathway inhibition promoted epithelial-related gene and protein expression. Furthermore, we demonstrated the ability of Epi-GMSCs to facilitate wound healing by improving re-epithelialization in a full-thickness skin defect model. CONCLUSIONS Collectively, this study uncovers that GMSCs have the ability to differentiate into epithelia and highlights a promising strategy for using Epi-GMSCs to improve cutaneous wound healing.
Collapse
Affiliation(s)
- Yongzheng Li
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, 310000, China
| | - Lingling Dong
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, 310000, China
| | - Yani Chen
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, 310000, China
| | - Wenjin Cai
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, 310000, China
| | - Guoli Yang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, 310000, China.
| | - Ying Wang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, 310000, China.
| |
Collapse
|
22
|
Chen C, Amona FM, Chen J, Chen X, Ke Y, Tang S, Xu J, Chen X, Pang Y. Multifunctional SEBS/AgNWs Nanocomposite Films with Antimicrobial, Antioxidant, and Anti-Inflammatory Properties Promote Infected Wound Healing. ACS APPLIED MATERIALS & INTERFACES 2024; 16:61751-61764. [PMID: 39479988 DOI: 10.1021/acsami.4c15649] [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: 11/02/2024]
Abstract
Wound healing is a complex biological process that can trigger inflammation and oxidative stress and impair myofibrillogenesis and angiogenesis. Several advanced wound-dressing nanocomposite materials have been designed to address these issues. Here, we designed a new multifunctional styrene-ethylene-butylene-styrene/silver nanowire (SEBS/AgNWs)-based nanocomposite film with antimicrobial, antioxidant, and anti-inflammatory properties to promote wound healing. The porous morphological structure of SEBS/AgNWs enhances their antimicrobial, antioxidant, and anti-inflammatory properties. SEBS/AgNWs significantly inhibited the growth of Staphylococcus aureus, methicillin-resistant Staphylococcus aureus, and Escherichia coli strains, effectively wiping out ABTS•+, DPPH•, hydrogen peroxide (H2O2), and hydroxyl (•OH) radicals, showing their effective ROS-scavenging properties. It further showed significant antioxidant properties by increasing the levels of enzyme-like catalase (CAT), superoxide dismutase (SOD), and glutathione (GSH), while decreasing malonaldehyde (MDA) levels. Additionally, SEBS/AgNWs reduced the expression of interleukin-1β (IL-1β), IL-6, and tumor necrosis factor-α (TNF-α), while increasing levels of transforming growth factor- β (TGF-β), vascular endothelial growth factor-A (VEGF), and CD31 in wound healing. This suggests that applying a multifunctional nanoplatform based on SEBS/AgNWs could enhance wound healing and improve patient outcomes in wound care management.
Collapse
Affiliation(s)
- Chen Chen
- College of Hydraulic Engineering Jiangsu Vocational Institute of Architectural Technology, Xuzhou 221000, China
- College of Water Resources and Hydropower Engineering, Yangzhou University, Yangzhou 225009, China
| | - Fructueux Modeste Amona
- Institute of Cellular and Molecular Biology, School of Life Science, Jiangsu Normal University, Xuzhou, Jiangsu 221116, China
| | - Junhao Chen
- Institute of Cellular and Molecular Biology, School of Life Science, Jiangsu Normal University, Xuzhou, Jiangsu 221116, China
| | - Xiaohan Chen
- Institute of Cellular and Molecular Biology, School of Life Science, Jiangsu Normal University, Xuzhou, Jiangsu 221116, China
| | - Yongding Ke
- Institute of Cellular and Molecular Biology, School of Life Science, Jiangsu Normal University, Xuzhou, Jiangsu 221116, China
| | - Shuangcheng Tang
- College of Water Resources and Hydropower Engineering, Yangzhou University, Yangzhou 225009, China
| | - Jinming Xu
- College of Water Resources and Hydropower Engineering, Yangzhou University, Yangzhou 225009, China
| | - Xi Chen
- Institute of Cellular and Molecular Biology, School of Life Science, Jiangsu Normal University, Xuzhou, Jiangsu 221116, China
| | - Yipeng Pang
- Institute of Cellular and Molecular Biology, School of Life Science, Jiangsu Normal University, Xuzhou, Jiangsu 221116, China
| |
Collapse
|
23
|
Ghasempour A, Dehghan H, Mahmoudi M, Lavi Arab F. Biomimetic scaffolds loaded with mesenchymal stem cells (MSCs) or MSC-derived exosomes for enhanced wound healing. Stem Cell Res Ther 2024; 15:406. [PMID: 39522032 PMCID: PMC11549779 DOI: 10.1186/s13287-024-04012-8] [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: 09/16/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024] Open
Abstract
Since wound healing is one of the most important medical challenges and common dressings have not been able to manage this challenge well today, efforts have been increased to achieve an advanced dressing. Mesenchymal stem cells and exosomes derived from them have shown high potential in healing and regenerating wounds due to their immunomodulatory, anti-inflammatory, immunosuppressive, and high regenerative capacities. However, challenges such as the short life of these cells, the low durability of these cells in the wound area, and the low stability of exosomes derived from them have resulted in limitations in their use for wound healing. Nowadays, different scaffolds are considered suitable biomaterials for wound healing. These scaffolds are made of natural or synthetic polymers and have shown promising potential for an ideal dressing that does not have the disadvantages of common dressings. One of the strategies that has attracted much attention today is using these scaffolds for seeding and delivering MSCs and their exosomes. This combined strategy has shown a high potential in enhancing the shelf life of cells and increasing the stability of exosomes. In this review, the combination of different scaffolds with different MSCs or their exosomes for wound healing has been comprehensively discussed.
Collapse
Affiliation(s)
- Alireza Ghasempour
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hamideh Dehghan
- Student Research Committee, Birjand University of Medical Sciences, Birjand, Iran
| | - Mahmoud Mahmoudi
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Immunology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Fahimeh Lavi Arab
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
- Department of Immunology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
24
|
Shi S, Ou X, Long J, Lu X, Xu S, Zhang L. Nanoparticle-Based Therapeutics for Enhanced Burn Wound Healing: A Comprehensive Review. Int J Nanomedicine 2024; 19:11213-11233. [PMID: 39513089 PMCID: PMC11542498 DOI: 10.2147/ijn.s490027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Accepted: 10/24/2024] [Indexed: 11/15/2024] Open
Abstract
Burn wounds pose intricate clinical challenges due to their severity and high risk of complications, demanding advanced therapeutic strategies beyond conventional treatments. This review discusses the application of nanoparticle-based therapies for optimizing burn wound healing. We explore the critical phases of burn wound healing, including inflammation, proliferation, and remodeling, while summarizing key nanoparticle-based strategies that influence these processes to optimize healing. Various nanoparticles, such as metal-based, polymer-based, and extracellular vesicles, are evaluated for their distinctive properties and mechanisms of action, including antimicrobial, anti-inflammatory, and regenerative effects. Future directions are highlighted, focusing on personalized therapies and the integration of sophisticated drug delivery systems, emphasizing the transformative potential of nanoparticles in enhancing burn wound treatment.
Collapse
Affiliation(s)
- Shaoyan Shi
- Department of Hand Surgery, Honghui Hospital, Xi’an Jiaotong University, Xi’an Honghui Hospital North District, Xi’an, Shaanxi, 710000, People’s Republic of China
| | - Xuehai Ou
- Department of Hand Surgery, Honghui Hospital, Xi’an Jiaotong University, Xi’an Honghui Hospital North District, Xi’an, Shaanxi, 710000, People’s Republic of China
| | - Jiafeng Long
- Department of Hand Surgery, Honghui Hospital, Xi’an Jiaotong University, Xi’an Honghui Hospital North District, Xi’an, Shaanxi, 710000, People’s Republic of China
| | - Xiqin Lu
- Department of Hand Surgery, Honghui Hospital, Xi’an Jiaotong University, Xi’an Honghui Hospital North District, Xi’an, Shaanxi, 710000, People’s Republic of China
| | - Siqi Xu
- Department of Hand Surgery, Honghui Hospital, Xi’an Jiaotong University, Xi’an Honghui Hospital North District, Xi’an, Shaanxi, 710000, People’s Republic of China
| | - Li Zhang
- Department of Hand Surgery, Honghui Hospital, Xi’an Jiaotong University, Xi’an Honghui Hospital North District, Xi’an, Shaanxi, 710000, People’s Republic of China
| |
Collapse
|
25
|
Wang F, Li S, Wang X, Yang Q, Duan J, Yang Y, Mu H. Gellan gum-based multifunctional hydrogel with enduring sterilization and ROS scavenging for infected wound healing. Int J Biol Macromol 2024; 282:136888. [PMID: 39490880 DOI: 10.1016/j.ijbiomac.2024.136888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 10/09/2024] [Accepted: 10/22/2024] [Indexed: 11/05/2024]
Abstract
The progression of severe skin injury healing can be easily impeded by bacterial infections and the resultant overproduction of reactive oxygen species (ROS) within the wound microenvironment. In this study, we developed a multifunctional antibacterial hydrogel by integrating gallium ion-tannic acid and polydopamine particles into gellan gum via a facile heat-cooling process. By harnessing the synergistic effects of polydopamine for short-term photothermal therapy and gallium ion for long-term chemotherapy, the hydrogel obtained shows outstanding antibacterial activities. Sustained release of gallium ion and tannic acid ensures a prolonged sterilization along with ROS-scavenging benefits. Moreover, this hydrogel demonstrates superior cytocompatibility, hemostatic properties, as well as capabilities including promoting cell migration, and adsorption to bacterial cells and toxin. The therapeutic efficacy of the hydrogel was validated using a mouse model of MRSA-induced cutaneous infections. Overall, this work introduces a straightforward yet highly efficient multifunctional hydrogel platform that combines synergetic antibacterial actions, ROS scavenging, and hemostasis to enhance the healing of bacteria-associated wounds.
Collapse
Affiliation(s)
- Fei Wang
- College of Chemistry & Pharmacy, Northwest A&F University, Yangling 712100, Shaanxi, China; Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi 830046, Xinjiang, China
| | - Siwei Li
- College of Chemistry & Pharmacy, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Xing Wang
- College of Chemistry & Pharmacy, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Qisen Yang
- College of Chemistry & Pharmacy, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Jinyou Duan
- College of Chemistry & Pharmacy, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Yu Yang
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi 830046, Xinjiang, China.
| | - Haibo Mu
- College of Chemistry & Pharmacy, Northwest A&F University, Yangling 712100, Shaanxi, China.
| |
Collapse
|
26
|
Tang S, Cai P, He H, Tian Y, Hao R, Liu X, Jing T, Xu Y, Li X. Global trends in the clinical utilization of exosomes in dermatology: a bibliometric analysis. Front Med (Lausanne) 2024; 11:1462085. [PMID: 39450105 PMCID: PMC11500466 DOI: 10.3389/fmed.2024.1462085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 09/11/2024] [Indexed: 10/26/2024] Open
Abstract
The arena of exosomal research presents substantial emerging prospects for clinical dermatology applications. This investigation conducts a thorough analysis of the contemporary global research landscape regarding exosomes and their implications for dermatological applications over the preceding decade. Employing bibliometric methodologies, this study meticulously dissects the knowledge framework and identifies dynamic trends within this specialized field. Contemporary scholarly literature spanning the last decade was sourced from the Web of Science Core Collection (WoSCC) database. Subsequent to retrieval, both quantitative and visual analyses of the pertinent publications were performed utilizing the analytical software tools VOSviewer and Citespace. A comprehensive retrieval yielded 545 scholarly articles dated from January 1, 2014, to December 31, 2023. Leading the research forefront are institutions such as Shanghai Jiao Tong University, The Fourth Military Medical University, and Sun Yat-sen University. The most prolific contributors on a national scale are China, the United States, and South Korea. Among the authors, Zhang Bin, Zhang Wei, and Zhang Yan emerge as the most published, with Zhang Bin also achieving the distinction of being the most cited. The International Journal of Molecular Sciences leads in article publications, whereas Stem Cell Research & Therapy holds the pinnacle in citation rankings. Theranostics boasts the highest impact factor among the periodicals. Current research hotspots in this area include Adipose mesenchymal stem cell-derived exosomes(ADSC-Exos), diabetic skin wounds, cutaneous angiogenesis, and the combination of biomaterials and exosomes. This manuscript constitutes the inaugural comprehensive bibliometric analysis that delineates the prevailing research trends and advancements in the clinical application of exosomes in dermatology. These analyses illuminate the contemporary research focal points and trajectories, providing invaluable insights that will inform further exploration within this domain.
Collapse
Affiliation(s)
- Shiqin Tang
- School of Clinical Medicine, Hebei University of Engineering, Handan, Hebei, China
| | - Pai Cai
- School of Information Engineering, Suihua University, Suihua, Heilongjiang, China
| | - Huina He
- School of Clinical Medicine, Hebei University of Engineering, Handan, Hebei, China
| | - Yanan Tian
- School of Clinical Medicine, Hebei University of Engineering, Handan, Hebei, China
| | - Ruiying Hao
- Affiliated Hospital of Hebei University of Engineering, Handan, Hebei, China
| | - Xin Liu
- Handan Stomatological Hospital, Endodontics, Handan, Hebei, China
| | - Tingting Jing
- Affiliated Hospital of Hebei University of Engineering, Handan, Hebei, China
- Hebei Key Laboratory of Immunological Dermatology, Handan, Hebei, China
| | - Yanyan Xu
- Affiliated Hospital of Hebei University of Engineering, Handan, Hebei, China
- Hebei Key Laboratory of Immunological Dermatology, Handan, Hebei, China
| | - Xiaojing Li
- School of Clinical Medicine, Hebei University of Engineering, Handan, Hebei, China
- Affiliated Hospital of Hebei University of Engineering, Handan, Hebei, China
- Hebei Key Laboratory of Immunological Dermatology, Handan, Hebei, China
| |
Collapse
|
27
|
Liu S, Zhao H, Jiang T, Wan G, Yan C, Zhang C, Yang X, Chen Z. The Angiogenic Repertoire of Stem Cell Extracellular Vesicles: Demystifying the Molecular Underpinnings for Wound Healing Applications. Stem Cell Rev Rep 2024; 20:1795-1812. [PMID: 39001965 DOI: 10.1007/s12015-024-10762-y] [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] [Accepted: 07/06/2024] [Indexed: 07/15/2024]
Abstract
Stem cells-derived extracellular vesicles (SC-EVs) have emerged as promising therapeutic agents for wound repair, recapitulating the biological effects of parent cells while mitigating immunogenic and tumorigenic risks. These EVs orchestrate wound healing processes, notably through modulating angiogenesis-a critical event in tissue revascularization and regeneration. This study provides a comprehensive overview of the multifaceted mechanisms underpinning the pro-angiogenic capacity of EVs from various stem cell sources within the wound microenvironment. By elucidating the molecular intricacies governing their angiogenic prowess, we aim to unravel the mechanistic repertoire underlying their remarkable potential to accelerate wound healing. Additionally, methods to enhance the angiogenic effects of SC-EVs, current limitations, and future perspectives are highlighted, emphasizing the significant potential of this rapidly advancing field in revolutionizing wound healing strategies.
Collapse
Affiliation(s)
- Shuoyuan Liu
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Huayuan Zhao
- Department of Urology, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Tao Jiang
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Gui Wan
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Chengqi Yan
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Chi Zhang
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xiaofan Yang
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Zhenbing Chen
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
28
|
Li Z, Li Q, Ahmad A, Yue Z, Wang H, Wu G. Highly concentrated collagen/chondroitin sulfate scaffold with platelet-rich plasma promotes bone-exposed wound healing in porcine. Front Bioeng Biotechnol 2024; 12:1441053. [PMID: 39380894 PMCID: PMC11458455 DOI: 10.3389/fbioe.2024.1441053] [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: 05/30/2024] [Accepted: 09/16/2024] [Indexed: 10/10/2024] Open
Abstract
In the case of wounds with exposed bone, it is essential to provide not only scaffolds with sufficient mechanical strength for protection, but also environments that are conducive to the regeneration of tissues and blood vessels. Despite the excellent biocompatibility and biodegradability of collagen and chondroitin sulfate, they display poor mechanical strength and rapid degradation rates. In contrast to previous methodologies that augmented the mechanical properties of biomaterials through the incorporation of additional substances, this investigation exclusively enhanced the mechanical strength of collagen/chondroitin sulfate scaffolds by modulating collagen concentrations. Furthermore, platelet-rich plasma (PRP) was employed to establish optimal conditions for vascular and tissue regeneration at the wound site. High-concentration collagen/chondroitin sulfate (H C-S) scaffolds were synthesized using high-speed centrifugation and combined with PRP, and their effects on endothelial cell proliferation were assessed. A porcine model of bone-exposed wounds was developed to investigate the healing effects and mechanisms. The experimental results indicated that scaffolds with increased collagen concentration significantly enhanced both tensile and compressive moduli. The combination of H C-S scaffolds with PRP markedly promoted endothelial cell proliferation. In vivo experiments demonstrated that this combination significantly accelerated the healing of porcine bone-exposed wounds and promoted vascular regeneration. This represents a promising strategy for promoting tissue regeneration that is worthy of further exploration and clinical application.
Collapse
Affiliation(s)
- Zhihao Li
- Department of Spinal Surgery, Jingzhou Hospital Affiliated to Yangtze University, Jingzhou, Hubei, China
| | - Qian Li
- Medical Laboratory of Jingzhou Hospital Affiliated to Yangtze University, Jingzhou, Hubei, China
| | - Akhlaq Ahmad
- Institutes of Integrative Medicine, Fudan University, Shanghai, China
| | - Zhongjie Yue
- Department of Spinal Surgery, Jingzhou Hospital Affiliated to Yangtze University, Jingzhou, Hubei, China
| | - Hongxia Wang
- Department of Spinal Surgery, Jingzhou Hospital Affiliated to Yangtze University, Jingzhou, Hubei, China
| | - Guofeng Wu
- Department of Orthopedics, Southern University of Science and Technology Hospital, Shenzhen, Guangdong, China
| |
Collapse
|
29
|
Xie P, Xue X, Li X. Recent Progress in Mesenchymal Stem Cell-Derived Exosomes for Skin Wound Repair. Cell Biochem Biophys 2024; 82:1651-1663. [PMID: 38811472 DOI: 10.1007/s12013-024-01328-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/18/2024] [Indexed: 05/31/2024]
Abstract
BACKGROUND Exosomes are nanometer-sized, lipid bilayer membrane vesicles that are secreted by various cell types. Mesenchymal stem cells (MSCs) have been shown to exert therapeutic effects through the secretion of exosomes via a paracrine pathway. Functions: Recent studies have demonstrated that MSC-derived exosomes (MSC-Exos) can effectively transport various bioactive substances, including proteins, mRNAs, microRNAs, long non-coding RNAs, circular RNAs, and lipids, into target cells. This process regulates multiple aspects during wound repair, such as the inflammatory response, cell proliferation, migration, differentiation, angiogenesis, and matrix remodeling. POTENTIAL APPLICATIONS By promoting wound healing and inhibiting scar formation, MSC-Exos have shown great promise for clinical applications in wound repair. This review highlights the recent advances in our understanding of the role and mechanism of MSC-Exos during wound repair, providing insights into their potential use in future therapeutic strategies.
Collapse
Affiliation(s)
- Peilin Xie
- Department of Plastic Surgery, People's Hospital of Gansu Province, Lanzhou, 730000, Gansu, China
| | - Xiaodong Xue
- Department of Plastic Surgery, People's Hospital of Gansu Province, Lanzhou, 730000, Gansu, China
| | - Xiaodong Li
- Center for Cosmetic Surgery, General Hospital of Lanzhou Petrochemical Company (The Fourth Affiliated Hospital of Gansu University of Chinese Medicine), Lanzhou, 730060, Gansu, China.
| |
Collapse
|
30
|
Qiu M, He Y, Zhang H, Zheng Y, Shi X, Yang J. Platelet-Rich Plasma (PRP) Based on Simple and Efficient Integrated Preparation Precises Quantitatively for Skin Wound Repair. Int J Mol Sci 2024; 25:9340. [PMID: 39273289 PMCID: PMC11394848 DOI: 10.3390/ijms25179340] [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: 07/03/2024] [Revised: 08/24/2024] [Accepted: 08/24/2024] [Indexed: 09/15/2024] Open
Abstract
Platelet-rich plasma (PRP) has become an important regenerative therapy. However, the preparation method of PRP has not been standardized, and the optimal platelet concentration for PRP used in skin wound repair is unclear, leading to inconsistent clinical efficacy of PRP. Therefore, the development of standardized preparation methods for PRP and the investigation of the dose-response relationship between PRP with different platelet concentrations and tissue regeneration plays an important role in the development and clinical application of PRP technology. This study has developed an integrated blood collection device from blood drawing to centrifugation. Response surface methodology was employed to optimize the preparation conditions, ultimately achieving a platelet recovery rate as high as 95.74% for PRP (with optimal parameters: centrifugation force 1730× g, centrifugation time 10 min, and serum separation gel dosage 1.4 g). Both in vitro and in vivo experimental results indicate that PRP with a (2×) enrichment ratio is the most effective in promoting fibroblast proliferation and skin wound healing, with a cell proliferation rate of over 150% and a wound healing rate of 78% on day 7.
Collapse
Affiliation(s)
- Mengjie Qiu
- College of Chemistry, Fuzhou University, No. 2 Xueyuan Road, Fuzhou 350108, China
- Fujian Key Laboratory of Medical Instrument and Pharmaceutical Technology, Fuzhou University, No. 2 Xueyuan Road, Fuzhou 350108, China
- International Joint Laboratory of Intelligent Health Care, Fuzhou University, No. 2 Xueyuan Road, Fuzhou 350108, China
| | - Yating He
- College of Chemistry, Fuzhou University, No. 2 Xueyuan Road, Fuzhou 350108, China
- Fujian Key Laboratory of Medical Instrument and Pharmaceutical Technology, Fuzhou University, No. 2 Xueyuan Road, Fuzhou 350108, China
- International Joint Laboratory of Intelligent Health Care, Fuzhou University, No. 2 Xueyuan Road, Fuzhou 350108, China
| | - Haijie Zhang
- College of Chemistry, Fuzhou University, No. 2 Xueyuan Road, Fuzhou 350108, China
- Fujian Key Laboratory of Medical Instrument and Pharmaceutical Technology, Fuzhou University, No. 2 Xueyuan Road, Fuzhou 350108, China
- International Joint Laboratory of Intelligent Health Care, Fuzhou University, No. 2 Xueyuan Road, Fuzhou 350108, China
| | - Yunquan Zheng
- College of Chemistry, Fuzhou University, No. 2 Xueyuan Road, Fuzhou 350108, China
- Fujian Key Laboratory of Medical Instrument and Pharmaceutical Technology, Fuzhou University, No. 2 Xueyuan Road, Fuzhou 350108, China
- International Joint Laboratory of Intelligent Health Care, Fuzhou University, No. 2 Xueyuan Road, Fuzhou 350108, China
| | - Xianai Shi
- Fujian Key Laboratory of Medical Instrument and Pharmaceutical Technology, Fuzhou University, No. 2 Xueyuan Road, Fuzhou 350108, China
- International Joint Laboratory of Intelligent Health Care, Fuzhou University, No. 2 Xueyuan Road, Fuzhou 350108, China
- College of Biological Science and Engineering, Fuzhou University, No. 2 Xueyuan Road, Fuzhou 350108, China
| | - Jianmin Yang
- Fujian Key Laboratory of Medical Instrument and Pharmaceutical Technology, Fuzhou University, No. 2 Xueyuan Road, Fuzhou 350108, China
- International Joint Laboratory of Intelligent Health Care, Fuzhou University, No. 2 Xueyuan Road, Fuzhou 350108, China
- College of Biological Science and Engineering, Fuzhou University, No. 2 Xueyuan Road, Fuzhou 350108, China
| |
Collapse
|
31
|
Fan MH, Pi JK, Zou CY, Jiang YL, Li QJ, Zhang XZ, Xing F, Nie R, Han C, Xie HQ. Hydrogel-exosome system in tissue engineering: A promising therapeutic strategy. Bioact Mater 2024; 38:1-30. [PMID: 38699243 PMCID: PMC11061651 DOI: 10.1016/j.bioactmat.2024.04.007] [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: 01/23/2024] [Revised: 03/24/2024] [Accepted: 04/08/2024] [Indexed: 05/05/2024] Open
Abstract
Characterized by their pivotal roles in cell-to-cell communication, cell proliferation, and immune regulation during tissue repair, exosomes have emerged as a promising avenue for "cell-free therapy" in clinical applications. Hydrogels, possessing commendable biocompatibility, degradability, adjustability, and physical properties akin to biological tissues, have also found extensive utility in tissue engineering and regenerative repair. The synergistic combination of exosomes and hydrogels holds the potential not only to enhance the efficiency of exosomes but also to collaboratively advance the tissue repair process. This review has summarized the advancements made over the past decade in the research of hydrogel-exosome systems for regenerating various tissues including skin, bone, cartilage, nerves and tendons, with a focus on the methods for encapsulating and releasing exosomes within the hydrogels. It has also critically examined the gaps and limitations in current research, whilst proposed future directions and potential applications of this innovative approach.
Collapse
Affiliation(s)
- Ming-Hui Fan
- Department of Orthopedic Surgery and Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, PR China
| | - Jin-Kui Pi
- Core Facilities, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, PR China
| | - Chen-Yu Zou
- Department of Orthopedic Surgery and Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, PR China
| | - Yan-Lin Jiang
- Department of Orthopedic Surgery and Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, PR China
| | - Qian-Jin Li
- Department of Orthopedic Surgery and Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, PR China
| | - Xiu-Zhen Zhang
- Department of Orthopedic Surgery and Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, PR China
| | - Fei Xing
- Department of Orthopedic Surgery and Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, PR China
| | - Rong Nie
- Department of Orthopedic Surgery and Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, PR China
| | - Chen Han
- Department of Orthopedic Surgery and Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, PR China
| | - Hui-Qi Xie
- Department of Orthopedic Surgery and Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, PR China
- Frontier Medical Center, Tianfu Jincheng Laboratory, Chengdu, Sichuan, 610212, PR China
| |
Collapse
|
32
|
Xu Y, Zhang Y, Tian H, Zhong Q, Yi K, Li F, Xue T, Wang H, Lao Y, Xu Y, Li Y, Long L, Li K, Tao Y, Li M. Smart Microneedle Arrays Integrating Cell-Free Therapy and Nanocatalysis to Treat Liver Fibrosis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2309940. [PMID: 38874114 PMCID: PMC11336984 DOI: 10.1002/advs.202309940] [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: 12/27/2023] [Revised: 06/02/2024] [Indexed: 06/15/2024]
Abstract
Liver fibrosis is a chronic pathological condition lacking specific clinical treatments. Stem cells, with notable potential in regenerative medicine, offer promise in treating liver fibrosis. However, stem cell therapy is hindered by potential immunological rejection, carcinogenesis risk, efficacy variation, and high cost. Stem cell secretome-based cell-free therapy offers potential solutions to address these challenges, but it is limited by low delivery efficiency and rapid clearance. Herein, an innovative approach for in situ implantation of smart microneedle (MN) arrays enabling precisely controlled delivery of multiple therapeutic agents directly into fibrotic liver tissues is developed. By integrating cell-free and platinum-based nanocatalytic combination therapy, the MN arrays can deactivate hepatic stellate cells. Moreover, they promote excessive extracellular matrix degradation by more than 75%, approaching normal levels. Additionally, the smart MN arrays can provide hepatocyte protection while reducing inflammation levels by ≈70-90%. They can also exhibit remarkable capability in scavenging almost 100% of reactive oxygen species and alleviating hypoxia. Ultimately, this treatment strategy can effectively restrain fibrosis progression. The comprehensive in vitro and in vivo experiments, supplemented by proteome and transcriptome analyses, substantiate the effectiveness of the approach in treating liver fibrosis, holding immense promise for clinical applications.
Collapse
Affiliation(s)
- Yanteng Xu
- Laboratory of Biomaterials and Translational MedicineCenter for Nanomedicine and Department of UltrasoundThe Third Affiliated HospitalSun Yat‐sen UniversityGuangzhou510630China
| | - Yixin Zhang
- Laboratory of Biomaterials and Translational MedicineCenter for Nanomedicine and Department of UltrasoundThe Third Affiliated HospitalSun Yat‐sen UniversityGuangzhou510630China
| | - Hao Tian
- Laboratory of Biomaterials and Translational MedicineCenter for Nanomedicine and Department of UltrasoundThe Third Affiliated HospitalSun Yat‐sen UniversityGuangzhou510630China
- Department of NeurologyThe Third Affiliated HospitalSun Yat‐sen UniversityGuangzhou510630China
| | - Qingguo Zhong
- Laboratory of Biomaterials and Translational MedicineCenter for Nanomedicine and Department of UltrasoundThe Third Affiliated HospitalSun Yat‐sen UniversityGuangzhou510630China
| | - Ke Yi
- Laboratory of Biomaterials and Translational MedicineCenter for Nanomedicine and Department of UltrasoundThe Third Affiliated HospitalSun Yat‐sen UniversityGuangzhou510630China
| | - Fenfang Li
- Laboratory of Biomaterials and Translational MedicineCenter for Nanomedicine and Department of UltrasoundThe Third Affiliated HospitalSun Yat‐sen UniversityGuangzhou510630China
| | - Tiantian Xue
- Laboratory of Biomaterials and Translational MedicineCenter for Nanomedicine and Department of UltrasoundThe Third Affiliated HospitalSun Yat‐sen UniversityGuangzhou510630China
| | - Haixia Wang
- Laboratory of Biomaterials and Translational MedicineCenter for Nanomedicine and Department of UltrasoundThe Third Affiliated HospitalSun Yat‐sen UniversityGuangzhou510630China
| | - Yeh‐Hsing Lao
- Department of Pharmaceutical SciencesUniversity at BuffaloThe State University of New YorkBuffaloNY14214USA
| | - Yingying Xu
- Center for Health ResearchGuangzhou Institutes of Biomedicine and HealthChinese Academy of SciencesGuangzhou510530China
- University of China Academy of SciencesBeijing100049China
| | - Yinxiong Li
- Center for Health ResearchGuangzhou Institutes of Biomedicine and HealthChinese Academy of SciencesGuangzhou510530China
- University of China Academy of SciencesBeijing100049China
| | - Ling Long
- Department of NeurologyThe Third Affiliated HospitalSun Yat‐sen UniversityGuangzhou510630China
| | - Kai Li
- Laboratory of Biomaterials and Translational MedicineCenter for Nanomedicine and Department of UltrasoundThe Third Affiliated HospitalSun Yat‐sen UniversityGuangzhou510630China
| | - Yu Tao
- Laboratory of Biomaterials and Translational MedicineCenter for Nanomedicine and Department of UltrasoundThe Third Affiliated HospitalSun Yat‐sen UniversityGuangzhou510630China
- Guangdong Provincial Key Laboratory of Liver DiseaseGuangzhou510630China
| | - Mingqiang Li
- Laboratory of Biomaterials and Translational MedicineCenter for Nanomedicine and Department of UltrasoundThe Third Affiliated HospitalSun Yat‐sen UniversityGuangzhou510630China
- Guangdong Provincial Key Laboratory of Liver DiseaseGuangzhou510630China
| |
Collapse
|
33
|
Hao R, Ye X, Chen X, Du J, Tian F, Zhang L, Ma G, Rao F, Xue J. Integrating Bioactive Graded Hydrogel with Radially Aligned Nanofibers to Dynamically Manipulate Wound Healing Process. ACS APPLIED MATERIALS & INTERFACES 2024; 16:37770-37782. [PMID: 38987992 DOI: 10.1021/acsami.4c09204] [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: 07/12/2024]
Abstract
Skin wound healing is a complex process that requires appropriate treatment and management. Using a single scaffold to dynamically manipulate angiogenesis, cell migration and proliferation, and tissue reconstruction during skin wound healing is a great challenge. We developed a hybrid scaffold platform that integrates the spatiotemporal delivery of bioactive cues with topographical cues to dynamically manipulate the wound-healing process. The scaffold comprised gelatin methacryloyl hydrogels and electrospun poly(ε-caprolactone)/gelatin nanofibers. The hydrogels had graded cross-linking densities and were loaded with two different functional bioactive peptides. The nanofibers comprised a radially aligned nanofiber array layer and a layer of random fibers. During the early stages of wound healing, the KLTWQELYQLKYKGI peptide, which mimics vascular endothelial growth factor, was released from the inner layer of the hydrogel to accelerate angiogenesis. During the later stages of wound healing, the IKVAVS peptide, which promotes cell migration, synergized with the radially aligned nanofiber membrane to promote cell migration, while the nanofiber membrane also supported further cell proliferation. In an in vivo rat skin wound-healing model, the hybrid scaffold significantly accelerated wound healing and collagen deposition, and the ratio of type I to type III collagen at the wound site resembled that of normal skin. The prepared scaffold dynamically regulated the skin tissue regeneration process in stages to achieve rapid wound repair with clinical application potential, providing a strategy for skin wound repair.
Collapse
Affiliation(s)
- Ruinan Hao
- State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029, P. R. China
- School of Medicine, South China University of Technology, Guangzhou 510006, China
| | - Xilin Ye
- State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029, P. R. China
| | - Xiaofeng Chen
- Trauma Center, Peking University People's Hospital, Beijing 100044, P.R. China
- Key Laboratory of Trauma and Neural Regeneration, Ministry of Education, National Trauma Medical Center, Peking University, Beijing 100044, P.R. China
| | - Jinzhi Du
- School of Medicine, South China University of Technology, Guangzhou 510006, China
| | - Feng Tian
- State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029, P. R. China
| | - Liqun Zhang
- State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029, P. R. China
| | - Guolin Ma
- Department of Radiology, China-Japan Friendship Hospital, Beijing 100029, P.R. China
| | - Feng Rao
- Trauma Center, Peking University People's Hospital, Beijing 100044, P.R. China
- Key Laboratory of Trauma and Neural Regeneration, Ministry of Education, National Trauma Medical Center, Peking University, Beijing 100044, P.R. China
| | - Jiajia Xue
- State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029, P. R. China
| |
Collapse
|
34
|
Souza ILM, Suzukawa AA, Josino R, Marcon BH, Robert AW, Shigunov P, Correa A, Stimamiglio MA. Cellular In Vitro Responses Induced by Human Mesenchymal Stem/Stromal Cell-Derived Extracellular Vesicles Obtained from Suspension Culture. Int J Mol Sci 2024; 25:7605. [PMID: 39062847 PMCID: PMC11277484 DOI: 10.3390/ijms25147605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 07/04/2024] [Accepted: 07/09/2024] [Indexed: 07/28/2024] Open
Abstract
Mesenchymal stem/stromal cells (MSCs) and their extracellular vesicles (MSC-EVs) have been described to have important roles in tissue regeneration, including tissue repair, control of inflammation, enhancing angiogenesis, and regulating extracellular matrix remodeling. MSC-EVs have many advantages for use in regeneration therapies such as facility for dosage, histocompatibility, and low immunogenicity, thus possessing a lower possibility of rejection. In this work, we address the potential activity of MSC-EVs isolated from adipose-derived MSCs (ADMSC-EVs) cultured on cross-linked dextran microcarriers, applied to test the scalability and reproducibility of EV production. Isolated ADMSC-EVs were added into cultured human dermal fibroblasts (NHDF-1), keratinocytes (HaCat), endothelial cells (HUVEC), and THP-1 cell-derived macrophages to evaluate cellular responses (i.e., cell proliferation, cell migration, angiogenesis induction, and macrophage phenotype-switching). ADMSC viability and phenotype were assessed during cell culture and isolated ADMSC-EVs were monitored by nanotracking particle analysis, electron microscopy, and immunophenotyping. We observed an enhancement of HaCat proliferation; NHDF-1 and HaCat migration; endothelial tube formation on HUVEC; and the expression of inflammatory cytokines in THP-1-derived macrophages. The increased expression of TGF-β and IL-1β was observed in M1 macrophages treated with higher doses of ADMSC-EVs. Hence, EVs from microcarrier-cultivated ADMSCs are shown to modulate cell behavior, being able to induce skin tissue related cells to migrate and proliferate as well as stimulate angiogenesis and cause balance between pro- and anti-inflammatory responses in macrophages. Based on these findings, we suggest that the isolation of EVs from ADMSC suspension cultures makes it possible to induce in vitro cellular responses of interest and obtain sufficient particle numbers for the development of in vivo concept tests for tissue regeneration studies.
Collapse
Affiliation(s)
- Ingrid L. M. Souza
- Laboratory of Basic Biology of Stem Cells (Labcet), Carlos Chagas Institute, Fiocruz, Curitiba 81350-010, PR, Brazil (A.A.S.); (B.H.M.); (A.W.R.); (P.S.)
| | - Andreia A. Suzukawa
- Laboratory of Basic Biology of Stem Cells (Labcet), Carlos Chagas Institute, Fiocruz, Curitiba 81350-010, PR, Brazil (A.A.S.); (B.H.M.); (A.W.R.); (P.S.)
| | - Raphaella Josino
- Albert Einstein Israelite Hospital, São Paulo 05652-900, SP, Brazil
| | - Bruna H. Marcon
- Laboratory of Basic Biology of Stem Cells (Labcet), Carlos Chagas Institute, Fiocruz, Curitiba 81350-010, PR, Brazil (A.A.S.); (B.H.M.); (A.W.R.); (P.S.)
- Confocal and Electronic Microscopy Facility (RPT07C), Carlos Chagas Institute, Fiocruz, Curitiba 81350-010, PR, Brazil
| | - Anny W. Robert
- Laboratory of Basic Biology of Stem Cells (Labcet), Carlos Chagas Institute, Fiocruz, Curitiba 81350-010, PR, Brazil (A.A.S.); (B.H.M.); (A.W.R.); (P.S.)
- Confocal and Electronic Microscopy Facility (RPT07C), Carlos Chagas Institute, Fiocruz, Curitiba 81350-010, PR, Brazil
| | - Patrícia Shigunov
- Laboratory of Basic Biology of Stem Cells (Labcet), Carlos Chagas Institute, Fiocruz, Curitiba 81350-010, PR, Brazil (A.A.S.); (B.H.M.); (A.W.R.); (P.S.)
| | - Alejandro Correa
- Laboratory of Basic Biology of Stem Cells (Labcet), Carlos Chagas Institute, Fiocruz, Curitiba 81350-010, PR, Brazil (A.A.S.); (B.H.M.); (A.W.R.); (P.S.)
| | - Marco A. Stimamiglio
- Laboratory of Basic Biology of Stem Cells (Labcet), Carlos Chagas Institute, Fiocruz, Curitiba 81350-010, PR, Brazil (A.A.S.); (B.H.M.); (A.W.R.); (P.S.)
| |
Collapse
|
35
|
Xia Y, Yan S, Wei H, Zhang H, Hou K, Chen G, Cao R, Zhu M. Multifunctional Porous Bilayer Artificial Skin for Enhanced Wound Healing. ACS APPLIED MATERIALS & INTERFACES 2024; 16:34578-34590. [PMID: 38946497 DOI: 10.1021/acsami.4c05074] [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: 07/02/2024]
Abstract
Meeting the exacting demands of wound healing encompasses rapid coagulation, superior exudate absorption, high antibacterial efficacy, and imperative support for cell growth. In this study, by emulating the intricate structure of natural skin, we prepare a multifunctional porous bilayer artificial skin to address these critical requirements. The bottom layer, mimicking the dermis, is crafted through freeze-drying a gel network comprising carboxymethyl chitosan (CMCs) and gelatin (GL), while the top layer, emulating the epidermis, is prepared via electrospinning poly(l-lactic acid) (PLLA) nanofibers. With protocatechuic aldehyde and gallium ion complexation (PA@Ga) as cross-linking agents, the bottom PA@Ga-CMCs/GL layer featured an adjustable pore size (78-138 μm), high hemostatic performance (67s), and excellent bacterial inhibition rate (99.9%), complemented by an impressive liquid-absorbing capacity (2000% swelling rate). The top PLLA layer, with dense micronanostructure and hydrophobic properties, worked as a shield to effectively thwarted liquid or bacterial penetration. Furthermore, accelerated wound closure, reduced inflammatory responses, and enhanced formation of hair follicles and blood vessels are achieved by the porous artificial skin covered on the surface of wound. Bilayer artificial skin integrates the advantages of nanofibers and freeze-drying porous materials to effectively replicate the protective properties of the epidermal layer of the skin, as well as the cell migration and tissue regeneration of the dermis. This bioabsorbable artificial skin demonstrates structural and functional comparability to real skin, which would advance the field of wound care through its multifaceted capabilities.
Collapse
Affiliation(s)
- Yuhan Xia
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Materials Science and Engineering, Donghua University, Shanghai 201620, China
| | - Sai Yan
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Materials Science and Engineering, Donghua University, Shanghai 201620, China
| | - Huidan Wei
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Materials Science and Engineering, Donghua University, Shanghai 201620, China
| | - Han Zhang
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Materials Science and Engineering, Donghua University, Shanghai 201620, China
| | - Kai Hou
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Materials Science and Engineering, Donghua University, Shanghai 201620, China
| | - Guoyin Chen
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Materials Science and Engineering, Donghua University, Shanghai 201620, China
| | - Ran Cao
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Materials Science and Engineering, Donghua University, Shanghai 201620, China
- Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, Donghua University, Shanghai 201620, P. R. China
| | - Meifang Zhu
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Materials Science and Engineering, Donghua University, Shanghai 201620, China
| |
Collapse
|
36
|
Jin X, Zhang J, Zhang Y, He J, Wang M, Hei Y, Guo S, Xu X, Liu Y. Different origin-derived exosomes and their clinical advantages in cancer therapy. Front Immunol 2024; 15:1401852. [PMID: 38994350 PMCID: PMC11236555 DOI: 10.3389/fimmu.2024.1401852] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Accepted: 06/13/2024] [Indexed: 07/13/2024] Open
Abstract
Exosomes, as a class of small extracellular vesicles closely related to the biological behavior of various types of tumors, are currently attracting research attention in cancer diagnosis and treatment. Regarding cancer diagnosis, the stability of their membrane structure and their wide distribution in body fluids render exosomes promising biomarkers. It is expected that exosome-based liquid biopsy will become an important tool for tumor diagnosis in the future. For cancer treatment, exosomes, as the "golden communicators" between cells, can be designed to deliver different drugs, aiming to achieve low-toxicity and low-immunogenicity targeted delivery. Signaling pathways related to exosome contents can also be used for safer and more effective immunotherapy against tumors. Exosomes are derived from a wide range of sources, and exhibit different biological characteristics as well as clinical application advantages in different cancer therapies. In this review, we analyzed the main sources of exosomes that have great potential and broad prospects in cancer diagnosis and therapy. Moreover, we compared their therapeutic advantages, providing new ideas for the clinical application of exosomes.
Collapse
Affiliation(s)
- Xiaoyan Jin
- Department of Cell Biology and Genetics, Medical College of Yan’an University, Yan’an, Shaanxi, China
| | - Jing Zhang
- Department of Cell Biology and Genetics, Medical College of Yan’an University, Yan’an, Shaanxi, China
- The Second Affiliated Hospital of Xi‘an Medical University, Xi’an, Shaanxi, China
| | - Yufu Zhang
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Yan’an University, Yan’an, Shaanxi, China
| | - Jing He
- Laboratory of Obstetrics and Gynecology, The Affiliated Hospital of Yan’an University, Yan’an, Shaanxi, China
| | - Mingming Wang
- Department of Cell Biology and Genetics, Medical College of Yan’an University, Yan’an, Shaanxi, China
| | - Yu Hei
- Department of Cell Biology and Genetics, Medical College of Yan’an University, Yan’an, Shaanxi, China
| | - Shutong Guo
- Department of Cell Biology and Genetics, Medical College of Yan’an University, Yan’an, Shaanxi, China
| | - Xiangrong Xu
- Department of Cell Biology and Genetics, Medical College of Yan’an University, Yan’an, Shaanxi, China
| | - Yusi Liu
- Department of Cell Biology and Genetics, Medical College of Yan’an University, Yan’an, Shaanxi, China
| |
Collapse
|
37
|
Yang S, Sun Y, Yan C. Recent advances in the use of extracellular vesicles from adipose-derived stem cells for regenerative medical therapeutics. J Nanobiotechnology 2024; 22:316. [PMID: 38844939 PMCID: PMC11157933 DOI: 10.1186/s12951-024-02603-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 05/28/2024] [Indexed: 06/09/2024] Open
Abstract
Adipose-derived stem cells (ADSCs) are a subset of mesenchymal stem cells (MSCs) isolated from adipose tissue. They possess remarkable properties, including multipotency, self-renewal, and easy clinical availability. ADSCs are also capable of promoting tissue regeneration through the secretion of various cytokines, factors, and extracellular vesicles (EVs). ADSC-derived EVs (ADSC-EVs) act as intercellular signaling mediators that encapsulate a range of biomolecules. These EVs have been found to mediate the therapeutic activities of donor cells by promoting the proliferation and migration of effector cells, facilitating angiogenesis, modulating immunity, and performing other specific functions in different tissues. Compared to the donor cells themselves, ADSC-EVs offer advantages such as fewer safety concerns and more convenient transportation and storage for clinical application. As a result, these EVs have received significant attention as cell-free therapeutic agents with potential future application in regenerative medicine. In this review, we focus on recent research progress regarding regenerative medical use of ADSC-EVs across various medical conditions, including wound healing, chronic limb ischemia, angiogenesis, myocardial infarction, diabetic nephropathy, fat graft survival, bone regeneration, cartilage regeneration, tendinopathy and tendon healing, peripheral nerve regeneration, and acute lung injury, among others. We also discuss the underlying mechanisms responsible for inducing these therapeutic effects. We believe that deciphering the biological properties, therapeutic effects, and underlying mechanisms associated with ADSC-EVs will provide a foundation for developing a novel therapeutic approach in regenerative medicine.
Collapse
Affiliation(s)
- Song Yang
- Institute of Life Sciences, Chongqing Medical University, Chongqing, 400016, People's Republic of China.
| | - Yiran Sun
- School of Pharmacy, Chengdu Medical College, Chengdu, 610500, People's Republic of China.
| | - Chenchen Yan
- School of Pharmacy, Chengdu Medical College, Chengdu, 610500, People's Republic of China
| |
Collapse
|
38
|
Papadopoulos KS, Piperi C, Korkolopoulou P. Clinical Applications of Adipose-Derived Stem Cell (ADSC) Exosomes in Tissue Regeneration. Int J Mol Sci 2024; 25:5916. [PMID: 38892103 PMCID: PMC11172884 DOI: 10.3390/ijms25115916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 05/24/2024] [Accepted: 05/27/2024] [Indexed: 06/21/2024] Open
Abstract
Adipose-derived stem cells (ADSCs) are mesenchymal stem cells with a great potential for self-renewal and differentiation. Exosomes derived from ADSCs (ADSC-exos) can imitate their functions, carrying cargoes of bioactive molecules that may affect specific cellular targets and signaling processes. Recent evidence has shown that ADSC-exos can mediate tissue regeneration through the regulation of the inflammatory response, enhancement of cell proliferation, and induction of angiogenesis. At the same time, they may promote wound healing as well as the remodeling of the extracellular matrix. In combination with scaffolds, they present the future of cell-free therapies and promising adjuncts to reconstructive surgery with diverse tissue-specific functions and minimal adverse effects. In this review, we address the main characteristics and functional properties of ADSC-exos in tissue regeneration and explore their most recent clinical application in wound healing, musculoskeletal regeneration, dermatology, and plastic surgery as well as in tissue engineering.
Collapse
Affiliation(s)
- Konstantinos S. Papadopoulos
- Department of Plastic and Reconstructive Surgery, 401 General Military Hospital of Athens, 11525 Athens, Greece;
- First Department of Pathology, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Christina Piperi
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 75 M. Asias Street, 11527 Athens, Greece
| | - Penelope Korkolopoulou
- First Department of Pathology, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| |
Collapse
|
39
|
Wu S, Sun S, Fu W, Yang Z, Yao H, Zhang Z. The Role and Prospects of Mesenchymal Stem Cells in Skin Repair and Regeneration. Biomedicines 2024; 12:743. [PMID: 38672102 PMCID: PMC11048165 DOI: 10.3390/biomedicines12040743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Revised: 03/01/2024] [Accepted: 03/11/2024] [Indexed: 04/28/2024] Open
Abstract
Mesenchymal stem cells (MSCs) have been recognized as a cell therapy with the potential to promote skin healing. MSCs, with their multipotent differentiation ability, can generate various cells related to wound healing, such as dermal fibroblasts (DFs), endothelial cells, and keratinocytes. In addition, MSCs promote neovascularization, cellular regeneration, and tissue healing through mechanisms including paracrine and autocrine signaling. Due to these characteristics, MSCs have been extensively studied in the context of burn healing and chronic wound repair. Furthermore, during the investigation of MSCs, their unique roles in skin aging and scarless healing have also been discovered. In this review, we summarize the mechanisms by which MSCs promote wound healing and discuss the recent findings from preclinical and clinical studies. We also explore strategies to enhance the therapeutic effects of MSCs. Moreover, we discuss the emerging trend of combining MSCs with tissue engineering techniques, leveraging the advantages of MSCs and tissue engineering materials, such as biodegradable scaffolds and hydrogels, to enhance the skin repair capacity of MSCs. Additionally, we highlight the potential of using paracrine and autocrine characteristics of MSCs to explore cell-free therapies as a future direction in stem cell-based treatments, further demonstrating the clinical and regenerative aesthetic applications of MSCs in skin repair and regeneration.
Collapse
Affiliation(s)
- Si Wu
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
- National Clinical Research Center for Digestive Diseases, Beijing 100050, China
| | - Shengbo Sun
- School of Basic Medical Sciences, Capital Medical University, Beijing 100050, China
| | - Wentao Fu
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
- National Clinical Research Center for Digestive Diseases, Beijing 100050, China
| | - Zhengyang Yang
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
- National Clinical Research Center for Digestive Diseases, Beijing 100050, China
| | - Hongwei Yao
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
- National Clinical Research Center for Digestive Diseases, Beijing 100050, China
| | - Zhongtao Zhang
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
- National Clinical Research Center for Digestive Diseases, Beijing 100050, China
| |
Collapse
|
40
|
Biniazan F, Stoian A, Haykal S. Adipose-Derived Stem Cells: Angiogenetic Potential and Utility in Tissue Engineering. Int J Mol Sci 2024; 25:2356. [PMID: 38397032 PMCID: PMC10889096 DOI: 10.3390/ijms25042356] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 02/07/2024] [Accepted: 02/13/2024] [Indexed: 02/25/2024] Open
Abstract
Adipose tissue (AT) is a large and important energy storage organ as well as an endocrine organ with a critical role in many processes. Additionally, AT is an enormous and easily accessible source of multipotent cell types used in our day for all types of tissue regeneration. The ability of adipose-derived stem cells (ADSCs) to differentiate into other types of cells, such as endothelial cells (ECs), vascular smooth muscle cells, or cardiomyocytes, is used in tissue engineering in order to promote/stimulate the process of angiogenesis. Being a key for future successful clinical applications, functional vascular networks in engineered tissue are targeted by numerous in vivo and ex vivo studies. The article reviews the angiogenic potential of ADSCs and explores their capacity in the field of tissue engineering (TE).
Collapse
Affiliation(s)
- Felor Biniazan
- Latner Thoracic Research Laboratories, Division of Thoracic Surgery, Toronto General Hospital Research Institute, University Health Network, 200 Elizabeth Street Suite 8N-869, Toronto, ON M5G2C4, Canada; (F.B.); (A.S.)
| | - Alina Stoian
- Latner Thoracic Research Laboratories, Division of Thoracic Surgery, Toronto General Hospital Research Institute, University Health Network, 200 Elizabeth Street Suite 8N-869, Toronto, ON M5G2C4, Canada; (F.B.); (A.S.)
| | - Siba Haykal
- Latner Thoracic Research Laboratories, Division of Thoracic Surgery, Toronto General Hospital Research Institute, University Health Network, 200 Elizabeth Street Suite 8N-869, Toronto, ON M5G2C4, Canada; (F.B.); (A.S.)
- Division of Plastic and Reconstructive Surgery, Department of Surgery, University of Toronto, 200 Elizabeth Street Suite 8N-869, Toronto, ON M5G2C4, Canada
| |
Collapse
|
41
|
Yao W, Song Z, Ma X, Huang Y, Zhang X, Li Y, Wei P, Zhang J, Xiong C, Yang S, Xu Y, Jing W, Zhao B, Zhang X, Han Y. Asymmetric adhesive SIS-based wound dressings for therapeutically targeting wound repair. J Nanobiotechnology 2024; 22:34. [PMID: 38238748 PMCID: PMC10797997 DOI: 10.1186/s12951-024-02294-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 01/02/2024] [Indexed: 01/22/2024] Open
Abstract
Severe tissue injuries pose a significant risk to human health. Conventional wound dressings fall short in achieving effective tissue regeneration, resulting in suboptimal postoperative healing outcomes. In this study, an asymmetric adhesive wound dressing (marked as SIS/PAA/LAP) was developed, originating from acrylate acid (AA) solution with laponite (LAP) nanoparticles polymerization and photo-crosslinked on the decellularized extracellular matrix small intestinal submucosa (SIS) patch. Extensive studies demonstrated that the SIS/PAA/LAP exhibited higher tissue adhesion strength (~ 33 kPa) and burst strength (~ 22 kPa) compared to conventional wound dressings like Tegaderm and tissue adhesive products. Importantly, it maintained favorable cell viability and demonstrated robust angiogenic capacity. In animal models of full-thickness skin injuries in rats and skin injuries in Bama miniature pigs, the SIS/PAA/LAP could be precisely applied to wound sites. By accelerating the formation of tissue vascularization, it displayed superior tissue repair outcomes. This asymmetrically adhesive SIS-based patch would hold promising applications in the field of wound dressings.
Collapse
Affiliation(s)
- Wende Yao
- School of Medicine, Nankai University, Tianjin, 300071, China
- Department of Plastic and Reconstructive Surgery, The First Medical Centre, Chinese PLA General Hospital, Beijing, 100853, China
| | - Zelong Song
- School of Medicine, Nankai University, Tianjin, 300071, China
- Department of Orthopaedics, The Fourth Medical Centre, Chinese PLA General Hospital, Beijing, 100048, China
| | - Xiaodong Ma
- Department of Neurosurgery, The First Medical Centre, Chinese PLA General Hospital, Beijing, 100048, China
| | - Yiqian Huang
- Beijing Biosis Healing Biological Technology Co., Ltd, Beijing, 102600, China
| | - Xueying Zhang
- Beijing Biosis Healing Biological Technology Co., Ltd, Beijing, 102600, China
| | - Yunhuan Li
- Beijing Biosis Healing Biological Technology Co., Ltd, Beijing, 102600, China
| | - Pengfei Wei
- Beijing Biosis Healing Biological Technology Co., Ltd, Beijing, 102600, China
| | - Julei Zhang
- Department of Plastic and Reconstructive Surgery, The First Medical Centre, Chinese PLA General Hospital, Beijing, 100853, China
- Department of Burn and Plastic Surgery, The 980st Hospital of the PLA Joint Logistics Support Force, Hebei, China
| | - Chenlu Xiong
- School of Medicine, Nankai University, Tianjin, 300071, China
- Department of Plastic and Reconstructive Surgery, The First Medical Centre, Chinese PLA General Hospital, Beijing, 100853, China
| | - Sihan Yang
- School of Medicine, Nankai University, Tianjin, 300071, China
- Department of Plastic and Reconstructive Surgery, The First Medical Centre, Chinese PLA General Hospital, Beijing, 100853, China
| | - Yujian Xu
- Department of Plastic and Reconstructive Surgery, The First Medical Centre, Chinese PLA General Hospital, Beijing, 100853, China
| | - Wei Jing
- Beijing Biosis Healing Biological Technology Co., Ltd, Beijing, 102600, China
| | - Bo Zhao
- Beijing Biosis Healing Biological Technology Co., Ltd, Beijing, 102600, China.
| | - Xuesong Zhang
- School of Medicine, Nankai University, Tianjin, 300071, China.
- Department of Orthopaedics, The Fourth Medical Centre, Chinese PLA General Hospital, Beijing, 100048, China.
| | - Yan Han
- School of Medicine, Nankai University, Tianjin, 300071, China.
- Department of Plastic and Reconstructive Surgery, The First Medical Centre, Chinese PLA General Hospital, Beijing, 100853, China.
| |
Collapse
|