1
|
Lin H, Zhou C, Li Q, Xie Q, Xia L, Liu L, Bao W, Xiong X, Zhang H, Zheng Z, Zhao J, Liang W. Nanotechnology-Assisted mesenchymal stem cells treatment for improved cartilage regeneration: A review of current practices. Biochem Pharmacol 2025; 237:116895. [PMID: 40154890 DOI: 10.1016/j.bcp.2025.116895] [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: 11/18/2024] [Revised: 02/26/2025] [Accepted: 03/24/2025] [Indexed: 04/01/2025]
Abstract
Cartilage tissue does not promptly elicit an inflammatory response upon injury, hence constraining its capacity for healing and self-regeneration. Mesenchymal Stem Cells (MSC) therapy, enhanced by nanotechnology, offers promising advancements in cartilage repair. Injuries to cartilage often cause chronic pain, where current treatments are inadequate. As MSCs can readily differentiate into chondrocytes and secrete soluble factors, they are essential components in tissue engineering of cartilage repair. Although, like other stem cell applications, clinical applications are restricted by poor post implantation survival and differentiation. Recent studies show that nanoparticles (NPs) can further improve MSC outcomes by promoting cell adhesion, and chondrogenic differentiation allowing for sustained growth factor release. In addition, nanomaterials can improve the biological activity of MSCs, by also facilitating the composition of a conducive microenvironment for cartilage repair. In this review, the application of nanofibrous scaffolds, hydrogels and nanoscale particulate matter to improve mechanical properties in cartilage tissue engineering, are discussed. Moreover, the MSCs and nanotechnology synergistic effects present hope of overcoming the limitations of conventional treatments. Nanotechnology greatly enhances the MSC based cartilage regeneration strategies and could provide better treatment for cartilage related diseases in the future. Future research should be aimed at standardizing MSC harvesting and culturing protocols and contrasting their long-term efficacy.
Collapse
Affiliation(s)
- Hongming Lin
- Department of Orthopedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan 316000 Zhejiang Province, China
| | - Chao Zhou
- Department of Orthopedics, Zhoushan Guanghua hospital, Zhoushan 316000 Zhejiang Province, China
| | - Qingping Li
- Medical Research Center, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan 316000 Zhejiang Province, China
| | - Qiong Xie
- Medical Research Center, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan 316000 Zhejiang Province, China
| | - Linying Xia
- Medical Research Center, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan 316000 Zhejiang Province, China
| | - Lu Liu
- Medical Research Center, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan 316000 Zhejiang Province, China
| | - Wenwen Bao
- Medical Research Center, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan 316000 Zhejiang Province, China
| | - Xiaochun Xiong
- Department of Orthopedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan 316000 Zhejiang Province, China
| | - Hao Zhang
- Department of Orthopedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan 316000 Zhejiang Province, China
| | - Zeping Zheng
- Department of Orthopedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan 316000 Zhejiang Province, China
| | - Jiayi Zhao
- Department of Orthopedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan 316000 Zhejiang Province, China.
| | - Wenqing Liang
- Department of Orthopedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan 316000 Zhejiang Province, China.
| |
Collapse
|
2
|
Li J, Liu L, Chen Y, Huang Y, Yang L. Exosomes derived from human umbilical cord mesenchymal stem cells attenuate senescence of peritoneal mesothelial cells by inhibiting oxidative stress. Int Immunopharmacol 2025; 158:114813. [PMID: 40354711 DOI: 10.1016/j.intimp.2025.114813] [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: 11/01/2024] [Revised: 05/04/2025] [Accepted: 05/05/2025] [Indexed: 05/14/2025]
Abstract
OBJECTIVE Aging is a natural process that affects cellular function. In peritoneal dialysis (PD), chronic exposure to dialysate induces oxidative stress (OS) in peritoneal mesothelial cells (PMCs), leading to cellular aging, fibrosis, and reduced dialysis efficacy. Mesenchymal stem cells (MSCs) have shown potential in alleviating cellular aging. This study investigates the role of exosomes (hUMSC-Exos) derived from human umbilical cord MSCs (hUMSCs) in mitigating PMC senescence and explores the underlying mechanisms. METHODS Human peritoneal mesothelial cells (HMrSV5) were cultured with 2.5 % glucose to induce senescence. Aging markers were assessed via Western blotting, β-galactosidase staining, and cell cycle analysis. hUMSC-Exos were characterized using Western blot, electron microscopy, and nanoparticle tracking analysis. Their uptake by HMrSV5 cells was confirmed through fluorescence microscopy. Various concentrations of hUMSC-Exos were tested, and OS levels were evaluated using reactive oxygen species (ROS), malondialdehyde (MDA), and superoxide dismutase (SOD) assays. The impact of the OS inhibitor N-acetyl-L-cysteine (NAC) on aging markers was also examined. RESULTS HMrSV5 cells treated with 2.5 % glucose exhibited increased expression of P53, P21, and P16, along with G0/G1 cell cycle arrest. Treatment with 150 μg/mL hUMSC-Exos reduced aging markers, decreased ROS and MDA levels, and increased SOD activity. Similar effects were observed with NAC treatment. CONCLUSION hUMSC-Exos alleviate PMCs aging by inhibiting OS, highlighting their potential to improve PD outcomes.
Collapse
Affiliation(s)
- Jia Li
- Departments of Geriatrics, The First Hospital of China Medical University, Shenyang, Liaoning 110001, PR China
| | - Lixin Liu
- Departments of Geriatrics, The First Hospital of China Medical University, Shenyang, Liaoning 110001, PR China
| | - Yiman Chen
- Departments of Geriatrics, The First Hospital of China Medical University, Shenyang, Liaoning 110001, PR China
| | - Yuling Huang
- Departments of Geriatrics, The First Hospital of China Medical University, Shenyang, Liaoning 110001, PR China
| | - Lina Yang
- Departments of Geriatrics, The First Hospital of China Medical University, Shenyang, Liaoning 110001, PR China; Department of International Physical Examination Center, The First Hospital of China Medical University, Shenyang, Liaoning 110001, PR China.
| |
Collapse
|
3
|
Khidr WA, Alfarttoosi KH, Taher WM, Alwan M, Ali Al-Nuaimi AM, Jawad MJ. A review of the role of tumor-derived exosomes in cancers treatment and progression. Int Immunopharmacol 2025; 157:114782. [PMID: 40334624 DOI: 10.1016/j.intimp.2025.114782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2025] [Revised: 04/23/2025] [Accepted: 04/29/2025] [Indexed: 05/09/2025]
Abstract
Tumor cells (TCs) produce exosomes (EXOs), nanovesicles formed in endosomes. Tumor-derived exosomes (TDEs) are tiny, bubble-shaped structures formed by TCs that include microRNAs (miRNA), proteins, enzymes, and copies of DNA and RNA. Many different kinds of cancer rely on TDEs. For instance, TDEs play a large role in the tumor microenvironment (TME) and promote tumor spread via many pathways. Furthermore, TDEs impact the efficacy of cancer treatments. Additionally, because of their low immunogenicity, high biocompatibility, and low toxicity, TDEs have been extensively used as drug delivery vehicles for cancer immunotherapy. Consequently, future cancer treatments may benefit from focusing on both the therapeutic function and the tumorigenic pathways of TDEs. Consequently, in this work, we have examined the roles of TDEs in cancer development, such as tumor angiogenesis, immune system evasion, and tumor metastasis. Then, we reviewed TDEs used to transport anticancer medicines, including chemotherapeutic medications, therapeutic compounds (including miRNA), and anticancer nanoparticles. We have concluded by outlining the challenges of clinical translation, including carcinogenicity and medication resistance, and by offering some suggestions for addressing these issues.
Collapse
Affiliation(s)
- Wajida Ataallah Khidr
- Department of Medical Laboratory Technics, College of Health and Medical Technology, Alnoor University, Mosul, Iraq
| | | | - Waam Mohammed Taher
- College of Nursing, National University of Science and Technology, Dhi Qar, Iraq
| | | | | | | |
Collapse
|
4
|
Fu Y, Qiu Z, Cao Y, Jiang M, Cui X. Hydrogel-exosome complexes: a novel strategy for cardiovascular regeneration. NANOSCALE 2025. [PMID: 40434070 DOI: 10.1039/d5nr00892a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2025]
Abstract
Cardiovascular disease (CVD) remains one of the leading causes of high mortality and morbidity worldwide, posing a substantial threat to global health. Mesenchymal stem cell (MSC) therapy has emerged as a promising treatment approach, primarily through the secretion of various bioactive factors. Exosomes (Exos), in particular, stand out as the most effective components, as their noncoding RNA and proteins play a crucial role in promoting the repair of cardiac function, positioning them a promising cell-free therapy for CVD. However, challenges such as poor stability, low delivery efficiency, weak targeting, and rapid immune-mediated clearance hinder the broader application of Exos, presenting significant obstacles for further clinical translation. Recent advancements in biomaterials, particularly hydrogels, offer new avenues for Exos-based CVD therapies. Hydrogels, with their ability to improve stability, release control, and targeting, have gained considerable attention in the biomedical field. This review explores the latest research developments regarding the treatment of CVD using Exos, and highlights their synergistic application with hydrogels, which provide valuable insights for advancing Exos-based therapies and developing novel therapeutic strategies for CVD.
Collapse
Affiliation(s)
- Yonglin Fu
- School of Basic Medical Sciences, Guangdong Medical University, Dongguan, 523808, China.
| | - Zixiong Qiu
- School of Basic Medical Sciences, Guangdong Medical University, Dongguan, 523808, China.
| | - Yifang Cao
- School of Basic Medical Sciences, Guangdong Medical University, Dongguan, 523808, China.
| | - Mei Jiang
- School of Basic Medical Sciences, Guangdong Medical University, Dongguan, 523808, China.
| | - Xiaojun Cui
- School of Basic Medical Sciences, Guangdong Medical University, Dongguan, 523808, China.
- Kashi University School of Medicine, Xinjiang, 844000, China
| |
Collapse
|
5
|
Mardi N, Khanicheragh P, Abbasi-Malati Z, Saghebasl S, Khosrowshahi ND, Chegeni SA, Javid F, Azari M, Salimi L, Rezabakhsh A, Milani SZ, Rahbarghazi R. Beneficial and challenges of exosome application in ischemic heart disease. Stem Cell Res Ther 2025; 16:247. [PMID: 40390086 PMCID: PMC12090443 DOI: 10.1186/s13287-025-04363-w] [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: 01/13/2025] [Accepted: 04/23/2025] [Indexed: 05/21/2025] Open
Abstract
Cardiovascular diseases are the main cause of death and disability in the clinical setting. Among several pathological conditions, myocardial infarction (MI) is a common clinical finding and happens due to the reduction or complete interruption of blood support. Stem cells and progenitors are valid cell sources with significant potential to alleviate several tissue injuries. Differentiation to mature and functional cells and the release of various growth factors, and cytokines are the main reparative mechanisms by which stem cells mediate their reparative tasks. Exosomes (Exos), a subset of extracellular vesicles (EVs), exhibit great theranostic potential in biomedicine. Along with whole-cell-based therapies, the pre-clinical and clinical application of Exos has been extended in animals and humans with ischemic heart diseases (IHD). Here, in this review article, we aimed to highlight the importance of Exos in IHD and address the mechanism of action by focusing on their regenerative potential.
Collapse
Affiliation(s)
- Narges Mardi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Parisa Khanicheragh
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Zahra Abbasi-Malati
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
- Tuberculosis and Lung Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Solmaz Saghebasl
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nafiseh Didar Khosrowshahi
- Stem Cell and Tissue Engineering Research Laboratory, Sahand University of Technology, Tabriz, 51335-1996, Iran
| | | | - Farzin Javid
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mahdiyeh Azari
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Leila Salimi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Aysa Rezabakhsh
- Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Soheil Zamen Milani
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Rahbarghazi
- Tuberculosis and Lung Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
- Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
6
|
Lei L, Zhou S, Zeng L, Gu Q, Xue H, Wang F, Feng J, Cui S, Shi L. Exosome-Based Therapeutics in Dermatology. Biomater Res 2025; 29:0148. [PMID: 40351703 PMCID: PMC12062580 DOI: 10.34133/bmr.0148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 01/13/2025] [Accepted: 01/25/2025] [Indexed: 05/14/2025] Open
Abstract
Exosomes (Exos) are tiny extracellular vesicles containing a variety of active biomolecules that play important parts in intercellular communication and influence the functions of target cells. The potential of Exos in the treatment of dermatological diseases has recently been well appreciated. This review highlights the constituents, function, and delivery of Exos, with a particular focus on their applications in skin therapy. Firstly, we offer a concise overview of the biochemical properties of Exos, including their sources, structures, and internal constituents. Subsequently, the biomedical functions of Exos and the latest advances in the extraction and purification of Exos are summarized. We further discuss the modes of delivery of Exos and underscore the potential of biomaterials in this regard. Finally, we summarize the application of Exo-aided therapy in dermatology. Overall, the objective of this review is to provide a comprehensive perspective on the applications and recent advancements of Exo-based approaches in treating skin diseases, with the intention of guiding future research efforts.
Collapse
Affiliation(s)
- Lanjie Lei
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Institute of Translational Medicine,
Zhejiang Shuren University, Hangzhou 310015, China
| | - Shaoyu Zhou
- The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou 325200, China
| | - Lingyao Zeng
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Institute of Translational Medicine,
Zhejiang Shuren University, Hangzhou 310015, China
| | - Qiancheng Gu
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Institute of Translational Medicine,
Zhejiang Shuren University, Hangzhou 310015, China
| | - Huaqian Xue
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Institute of Translational Medicine,
Zhejiang Shuren University, Hangzhou 310015, China
| | - Fangyan Wang
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Institute of Translational Medicine,
Zhejiang Shuren University, Hangzhou 310015, China
| | - Jiayin Feng
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Institute of Translational Medicine,
Zhejiang Shuren University, Hangzhou 310015, China
| | - Shumao Cui
- School of Food Science and Technology,
Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Liyun Shi
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Institute of Translational Medicine,
Zhejiang Shuren University, Hangzhou 310015, China
| |
Collapse
|
7
|
Liu N, Wu T, Han G, Chen M. Exosome-mediated ferroptosis in the tumor microenvironment: from molecular mechanisms to clinical application. Cell Death Discov 2025; 11:221. [PMID: 40328736 PMCID: PMC12056189 DOI: 10.1038/s41420-025-02484-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2025] [Revised: 04/01/2025] [Accepted: 04/07/2025] [Indexed: 05/08/2025] Open
Abstract
Ferroptosis in the tumor microenvironment (TME) plays a crucial role in the development, metastasis, immune escape, and drug resistance of various types of cancer. A better understanding of ferroptosis in the TME could illuminate novel aspects of this process and promote the development of targeted therapies. Compelling evidence indicates that exosomes are key mediators in regulating the TME. In this respect, it is now understood that exosomes can deliver biologically functional molecules to recipient cells, influencing cancer progression by reprogramming the metabolism of cancer cells and their surrounding stromal cells through ferroptosis. In this review, we focus on the role of exosomes in the TME and describe how they contribute to tumor reprogramming, immunosuppression, and the formation of pre-metastatic niches through ferroptosis. In addition, we highlight exosome-mediated ferroptosis as a potential target for cancer therapy and discuss strategies employing exosomes in ferroptosis treatment. Finally, we outline the current applications and challenges of targeted exosome-mediated ferroptosis therapy in tumor immunotherapy and chemotherapy. Our aim is to advance research on the link between exosomes and ferroptosis in the TME, and we pose questions to guide future studies in this area.
Collapse
Affiliation(s)
- Na Liu
- Department of Radiotherapy and Oncology, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China
| | - Tianqing Wu
- XJTLU Wisdom Lake Academy of Pharmacy, Suzhou, Jiangsu Province, China
| | - Guohu Han
- Department of Oncology, Jingjiang People's Hospital Affiliated with Yangzhou University, Jingjiang, China
| | - Minbin Chen
- Department of Radiotherapy and Oncology, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China.
| |
Collapse
|
8
|
Ajwad N, Mustapha M, Idris Z, Lee SY. The Recent Applications of Stem Cell-Derived Exosomes and Hydrogels in Neurological Disorders. TISSUE ENGINEERING. PART B, REVIEWS 2025. [PMID: 40323680 DOI: 10.1089/ten.teb.2024.0353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/07/2025]
Abstract
Neurological disorders such as Alzheimer's disease, Parkinson's disease, and stroke pose significant challenges for conventional therapy due to the complexities of the blood-brain barrier (BBB) and the restricted delivery of drugs to the central nervous system. Exosomes, a type of small extracellular vesicle secreted by nearly all cell types, hold substantial promise as delivery vehicles for therapeutic agents in treating these conditions. Notably, stem cell-secreted exosomes have emerged as particularly effective due to their regenerative potential and natural ability to cross the BBB. Similarly, hydrogels have gained recognition as versatile biomaterials capable of supporting sustained release and targeted delivery of therapeutics. The combination of the regenerative properties of stem cell-derived exosomes (SC-Exos) with the structural and functional benefits of hydrogels offers a promising approach for enhancing neurogenesis, modulating neuroinflammation, and facilitating tissue repair. This review explores the origin, structure, and modifications of exosomes as well as the synthesis and incorporation methods of hydrogels in the therapeutic context for debilitating neurological disorders. It highlights recent advancements in using SC-Exos and hydrogels for therapeutic delivery, addressing both current challenges and future applications. Improving our understanding of hydrogels loaded with SC-Exos for cargo transportation and neural tissue regeneration may pave the way for novel therapeutic strategies.
Collapse
Affiliation(s)
- Nabil Ajwad
- Regenerative Medicine Research Group, Department of Hematology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Malaysia
| | - Muzaimi Mustapha
- Department of Neurosciences, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Malaysia
| | - Zamzuri Idris
- Department of Neurosciences, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Malaysia
| | - Si-Yuen Lee
- Regenerative Medicine Research Group, Department of Hematology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Malaysia
| |
Collapse
|
9
|
Ye YM, Zhao YX, Xiang LR, Zou CY, Xiao H, Lu H, Yang H, Hu JJ, Xie HQ. The Immunomodulatory mechanism and research progress of mesenchymal stem cells in the treatment of allergic rhinitis. Stem Cell Res Ther 2025; 16:188. [PMID: 40251675 PMCID: PMC12008879 DOI: 10.1186/s13287-025-04333-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2025] [Accepted: 04/10/2025] [Indexed: 04/20/2025] Open
Abstract
BACKGROUND Allergic rhinitis (AR) affects 10-40% of the global population, yet current therapies (drugs, immunotherapy) face limitations in efficacy and safety. Mesenchymal stem cells (MSCs) have emerged as a promising alternative due to their immunomodulatory properties. KEY FINDINGS Preclinical studies demonstrate that MSCs from adipose, bone marrow, umbilical cord, and tonsils reduce AR symptoms (sneezing, nasal inflammation) and serum IgE (Immunoglobulin E) levels by restoring Th1/Th2 immune equilibrium and enhancing Treg (Regulatory T cells) activity. MSC-derived exosomes and hydrogel-encapsulated formulations further improve targeting and safety. However, clinical translation is hindered by heterogeneous protocols and unresolved long-term risks (e.g., tumorigenicity). CLINICAL SIGNIFICANCE MSC-based therapies offer potential for durable AR remission by addressing immune dysregulation at its root. Future efforts must prioritize standardized production, phase I safety trials, and combination strategies (e.g., exosomes + hydrogels) to accelerate clinical adoption.
Collapse
Affiliation(s)
- Yu-Meng Ye
- Department of Otolaryngology-Head & Neck Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, P. R. China
- Department of Orthopedic Surgery and Orthopedic Research Institute, Stem Cell and Tissue Engineering Research Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, P. R. China
| | - Yu-Xin Zhao
- Department of Otolaryngology-Head & Neck Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, P. R. China
| | - Li-Rong Xiang
- Department of Otolaryngology-Head & Neck Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, P. R. China
| | - Chen-Yu Zou
- Department of Orthopedic Surgery and Orthopedic Research Institute, Stem Cell and Tissue Engineering Research Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, P. R. China
| | - Hao Xiao
- Department of Otolaryngology-Head & Neck Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, P. R. China
| | - Huan Lu
- Department of Otolaryngology-Head & Neck Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, P. R. China
- Department of Otolaryngology-Head & Neck Surgery, West China Tianfu Hospital, Sichuan University, Chengdu, Sichuan, 610213, P. R. China
| | - Hui Yang
- Department of Otolaryngology-Head & Neck Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, P. R. China
| | - Juan-Juan Hu
- Department of Otolaryngology-Head & Neck Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, P. R. China.
- Department of Orthopedic Surgery and Orthopedic Research Institute, Stem Cell and Tissue Engineering Research Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, P. R. China.
| | - Hui-Qi Xie
- Department of Orthopedic Surgery and Orthopedic Research Institute, Stem Cell and Tissue Engineering Research Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, P. R. China.
| |
Collapse
|
10
|
Shen Z, Shan Y, Lu Y, Zhu J, Yuan L, Chen W, Sun F, Wang Q, Wang Y, Zhang Y, Xu X, Chen Y, Ge W, Chen W, Si P, Zhang R, Shi H. Decellularized extracellular matrix-loaded exosome hydrogel for cell-free tracheal scaffold in tracheal defect reconstruction and repair. J Nanobiotechnology 2025; 23:289. [PMID: 40229874 PMCID: PMC11998422 DOI: 10.1186/s12951-025-03328-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 03/13/2025] [Indexed: 04/16/2025] Open
Abstract
Tracheal reconstruction presents a significant global clinical challenge due to the unique structure and function of the trachea, which complicates its repair. Key challenges include restoring tracheal function post-transplantation, managing surgical complications, and ensuring the long-term survival of transplanted tissue. Furthermore, adequate vascularization of the transplanted trachea, along with the repair of cartilage and epithelial cells, is critical for facilitating functional recovery and successful reconstruction. In this study, a decellularized tracheal matrix was extracted to preserve its natural bioactivity. Nanoclay and GelMA were then employed as carrier materials to integrate with the decellularized tracheal matrix, forming a hydrogel scaffold. Both nanoclay and GelMA offer tunable porous structures and surface properties that promote cell adhesion and proliferation, while providing sufficient mechanical support. By leveraging the biological functions of endothelial progenitor cell-derived exosomes, we successfully loaded exosomes onto the decellularized extracellular matrix, creating a novel cell-free tracheal scaffold for investigating tracheal defect repair. The scaffold exhibited excellent biocompatibility, promoting graft vascularization and facilitating the repair of tracheal cartilage and epithelium. These findings hold significant promise for advancing tracheal reconstruction surgery and offer valuable insights for future research in this area.
Collapse
Affiliation(s)
- Zhiming Shen
- Department of Thoracic Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230001, China
- Medical College, Yangzhou University, Yangzhou, 225009, China
| | - Yibo Shan
- Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou University, Yangzhou, 225001, China
- Medical College, Yangzhou University, Yangzhou, 225009, China
| | - Yi Lu
- Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou University, Yangzhou, 225001, China
- Medical College, Yangzhou University, Yangzhou, 225009, China
| | - Jianwei Zhu
- Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou University, Yangzhou, 225001, China
- Medical College, Yangzhou University, Yangzhou, 225009, China
| | - Lei Yuan
- Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou University, Yangzhou, 225001, China
- Medical College, Yangzhou University, Yangzhou, 225009, China
| | - Wenxuan Chen
- Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou University, Yangzhou, 225001, China
- Medical College, Yangzhou University, Yangzhou, 225009, China
| | - Fei Sun
- Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou University, Yangzhou, 225001, China
- Medical College, Yangzhou University, Yangzhou, 225009, China
| | - Qi Wang
- Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou University, Yangzhou, 225001, China
- Medical College, Yangzhou University, Yangzhou, 225009, China
| | - Yilun Wang
- Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou University, Yangzhou, 225001, China
- Medical College, Yangzhou University, Yangzhou, 225009, China
| | - Yaojing Zhang
- Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou University, Yangzhou, 225001, China
- Medical College, Yangzhou University, Yangzhou, 225009, China
| | - Xiangyu Xu
- Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou University, Yangzhou, 225001, China
- Medical College, Yangzhou University, Yangzhou, 225009, China
| | - Yu Chen
- Department of Thoracic Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230001, China
| | - Wei Ge
- Department of Thoracic Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230001, China
| | - Wei Chen
- Department of Thoracic Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230001, China
| | - Panpan Si
- Department of Thoracic Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230001, China
| | - Renquan Zhang
- Department of Thoracic Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230001, China
| | - Hongcan Shi
- Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou University, Yangzhou, 225001, China.
- Medical College, Yangzhou University, Yangzhou, 225009, China.
| |
Collapse
|
11
|
Nochalabadi A, Khazaei M, Rezakhani L. Exosomes and tissue engineering: A novel therapeutic strategy for nerve regenerative. Tissue Cell 2025; 93:102676. [PMID: 39693896 DOI: 10.1016/j.tice.2024.102676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 11/10/2024] [Accepted: 12/06/2024] [Indexed: 12/20/2024]
Abstract
Damage to nerves negatively impacts quality of life and causes considerable morbidity. Self-regeneration is a special characteristic of the nervous system, yet how successful regeneration is accomplished remains unclear. Research on nerve regeneration is advancing and accelerating successful nerve recovery with potential new approaches. Eukaryote cells release extracellular vesicles (EVs), which control intercellular communication in both health and disease. More and more, EVs such as microvesicles and exosomes (EXOs) are being recognized as viable options for cell-free therapies that address complex tissue regeneration. The present study highlights the functional relevance of EVs in regenerative medicine for nerve-related regeneration. A subclass of EVs, EXOs were first identified as a way for cells to expel undesirable cell products. These nanovesicles have a diameter of 30-150 nm and are secreted by a variety of cells in conditions of both health and illness. Their benefits include the ability to promote endothelial cell growth, inhibit inflammation, encourage cell proliferation, and regulate cell differentiation. They are also known to transport functional proteins, metabolites, and nucleic acids to recipient cells, thus playing a significant role in cellular communication. EXOs impact an extensive array of physiological functions, including immunological responses, tissue regeneration, stem cell conservation, communication within the central nervous system, and pathological processes involving cardiovascular disorders, neurodegeneration, cancer, and inflammation. Their biocompatibility and bi-layered lipid structure (which shields the genetic consignment from deterioration and reduces immunogenicity) make them appealing as therapeutic vectors. They can pass through the blood brain barrier and other major biological membranes because of their small size and membrane composition. The creation of modified EXOs is a dynamic area of research that supports the evaluation of diverse therapeutic freights, improvement of target selectivity, and manufacturing optimization.
Collapse
Affiliation(s)
- Azadeh Nochalabadi
- Fertility and Infertility Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mozafar Khazaei
- Fertility and Infertility Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran; Department of Tissue Engineering, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| | - Leila Rezakhani
- Fertility and Infertility Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran; Department of Tissue Engineering, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| |
Collapse
|
12
|
Daghrery A, Araújo IJDS, Marques JF, Alipour M, Ünsal RBK, Chathoth BM, Sivaramakrishnan G, Delgadillo-Barrera S, Chaurasia A. Role of exosomes in dental and craniofacial regeneration - A review. Tissue Cell 2025; 93:102684. [PMID: 39740273 DOI: 10.1016/j.tice.2024.102684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 12/05/2024] [Accepted: 12/11/2024] [Indexed: 01/02/2025]
Abstract
BACKGROUND The treatment of congenital deformities, traumatic injuries, infectious diseases, and tumors in the craniomaxillofacial (CMF) region is complex due to the intricate nature of the tissues involved. Conventional treatments such as bone grafts and cell transplantation face limitations, including the need for multiple surgeries, complications, and safety concerns. OBJECTIVE This paper aims to provide a comprehensive analysis of the role of exosomes (EXOs) in CMF and dental tissue regeneration and to explore their potential applications in regenerative dental medicine. METHODS An extensive review of advancements in tissue engineering, materials sciences, and nanotechnology was conducted to evaluate the development of delivery systems for EXOs-based therapies. The analysis included how EXOs, as nanovesicles released by cells, can be modified to target specific cells or loaded with functional molecules for drug or gene delivery. RESULTS EXOs have emerged as a promising alternative to cell transplant therapy, offering a safer method for cell communication and epigenetic control. EXOs transport important proteins and genetic materials, facilitating intercellular communication and delivering therapeutics effectively. The potential of EXOs in personalized medicine, particularly in diagnosing, customizing treatment, and predicting patient responses, is highlighted. CONCLUSION EXO-mediated therapy holds significant potential for advancing tissue regeneration, offering targeted, personalized treatment options with reduced side effects. However, challenges in purification, production, and standardized protocols need to be addressed before its clinical application can be fully realized.
Collapse
Affiliation(s)
- Arwa Daghrery
- Department of Restorative Dental Sciences, School of Dentistry, Jazan University, Jazan, Kingdom of Saudi Arabia.
| | | | - Joana Faria Marques
- Faculdade de Medicina Dentária, Universidade de Lisboa, Cidade Universitária, Lisboa 1600-277, Portugal.
| | - Mahdieh Alipour
- Dental and Periodontal Research Center, Faculty of Dentistry, Tabriz University of Medical Sciences, Iran; Departments of Oral and Craniofacial Sciences, School of Dental Medicine, University of Pittsburgh, USA.
| | - Revan Birke Koca Ünsal
- Department of Periodontology, University of Kyrenia, Faculty of Dentistry, Kyrenia, Cyprus.
| | | | | | - Sara Delgadillo-Barrera
- Grupo de Investigacion Básica y Aplicada en Odontología - IBAPO, Facultad de Odontologia, Universidad Nacional de Colombia, Bogotá, Colombia.
| | - Akhilanand Chaurasia
- Department of Oral Medicine and Radiology, Faculty of Dental Sciences. King George's Medical University, Lucknow, India.
| |
Collapse
|
13
|
Geng JX, Lu YF, Zhou JN, Huang B, Qin Y. Exosome technology: A novel and effective drug delivery system in the field of cancer therapy. World J Gastrointest Oncol 2025; 17:101857. [PMID: 40092946 PMCID: PMC11866225 DOI: 10.4251/wjgo.v17.i3.101857] [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: 09/28/2024] [Revised: 11/23/2024] [Accepted: 12/20/2024] [Indexed: 02/14/2025] Open
Abstract
In this article, we revisit an article, which specifically focuses on the utilization of exosomes derived from human bone marrow mesenchymal stem cells (MSCs) for targeted delivery of gemcitabine in pancreatic cancer treatment. The experimental results demonstrated that the exosome-based drug delivery system derived from MSCs significantly augmented apoptosis in pancreatic cancer cells. The biocompatibility, targeting specificity, and low immunogenicity of exosomes render them as optimal carriers for drug delivery, enabling precise administration of therapeutics to diseased tissues while mitigating adverse effects, thereby achieving targeted treatment of cancer cells and significantly enhancing anti-tumor efficacy. However, the clinical application of exosome drug delivery platforms in oncology still presents challenges, necessitating further optimization to ensure their stability and efficacy. This study focuses on elucidating the advantages of exosomes as a drug delivery platform, exploring the utilization of MSC-derived exosomes in oncology therapy, and discussing their potential and future directions in cancer treatment.
Collapse
Affiliation(s)
- Jia-Xin Geng
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, Zhejiang Province, China
| | - Yao-Fan Lu
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, Zhejiang Province, China
| | - Jing-Nan Zhou
- Zhejiang Cancer Hospital, Hangzhou 310018, Zhejiang Province, China
| | - Biao Huang
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, Zhejiang Province, China
| | - Yuan Qin
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, Zhejiang Province, China
| |
Collapse
|
14
|
Odehnalová N, Šandriková V, Hromadka R, Skaličková M, Dytrych P, Hoskovec D, Kejík Z, Hajduch J, Vellieux F, Vašáková MK, Martásek P, Jakubek M. The potential of exosomes in regenerative medicine and in the diagnosis and therapies of neurodegenerative diseases and cancer. Front Med (Lausanne) 2025; 12:1539714. [PMID: 40182844 PMCID: PMC11966052 DOI: 10.3389/fmed.2025.1539714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 02/06/2025] [Indexed: 04/05/2025] Open
Abstract
Exosomes, nanosized extracellular vesicles released by various cell types, are intensively studied for the diagnosis and treatment of cancer and neurodegenerative diseases, and they also display high usability in regenerative medicine. Emphasizing their diagnostic potential, exosomes serve as carriers of disease-specific biomarkers, enabling non-invasive early detection and personalized medicine. The cargo loading of exosomes with therapeutic agents presents an innovative strategy for targeted drug delivery, minimizing off-target effects and optimizing therapeutic interventions. In regenerative medicine, exosomes play a crucial role in intercellular communication, facilitating tissue regeneration through the transmission of bioactive molecules. While acknowledging existing challenges in standardization and scalability, ongoing research efforts aim to refine methodologies and address regulatory considerations. In summary, this review underscores the transformative potential of exosomes in reshaping the landscape of medical interventions, with a particular emphasis on cancer, neurodegenerative diseases, and regenerative medicine.
Collapse
Affiliation(s)
- Nikola Odehnalová
- NEXARS Research and Development Center C2P s.r.o, Chlumec nad Cidlinou, Czechia
- BIOCEV, First Faculty of Medicine, Charles University, Vestec, Czechia
| | - Viera Šandriková
- NEXARS Research and Development Center C2P s.r.o, Chlumec nad Cidlinou, Czechia
| | - Róbert Hromadka
- NEXARS Research and Development Center C2P s.r.o, Chlumec nad Cidlinou, Czechia
- BIOCEV, First Faculty of Medicine, Charles University, Vestec, Czechia
| | - Markéta Skaličková
- BIOCEV, First Faculty of Medicine, Charles University, Vestec, Czechia
- Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czechia
| | - Petr Dytrych
- Department of Surgery-Department of Abdominal, Thoracic Surgery and Traumatology, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czechia
| | - David Hoskovec
- Department of Surgery-Department of Abdominal, Thoracic Surgery and Traumatology, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czechia
| | - Zdeněk Kejík
- BIOCEV, First Faculty of Medicine, Charles University, Vestec, Czechia
- Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czechia
- Department of Analytical Chemistry, University of Chemistry and Technology, Prague, Czechia
| | - Jan Hajduch
- BIOCEV, First Faculty of Medicine, Charles University, Vestec, Czechia
- Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czechia
- The Department of Chemistry of Natural Compounds, University of Chemistry and Technology, Prague, Czechia
| | - Frédéric Vellieux
- BIOCEV, First Faculty of Medicine, Charles University, Vestec, Czechia
- Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czechia
| | - Martina Koziar Vašáková
- Department of Respiratory Medicine, First Faculty of Medicine, Charles University and Thomayer Hospital, Prague, Czechia
| | - Pavel Martásek
- Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czechia
| | - Milan Jakubek
- BIOCEV, First Faculty of Medicine, Charles University, Vestec, Czechia
- Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czechia
- Department of Analytical Chemistry, University of Chemistry and Technology, Prague, Czechia
| |
Collapse
|
15
|
Camacho-Cardenosa M, Pulido-Escribano V, Estrella-Guisado G, Dorado G, Herrera-Martínez AD, Gálvez-Moreno MÁ, Casado-Díaz A. Bioprinted Hydrogels as Vehicles for the Application of Extracellular Vesicles in Regenerative Medicine. Gels 2025; 11:191. [PMID: 40136896 PMCID: PMC11941778 DOI: 10.3390/gels11030191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Revised: 03/06/2025] [Accepted: 03/07/2025] [Indexed: 03/27/2025] Open
Abstract
Three-dimensional bioprinting is a new advance in tissue engineering and regenerative medicine. Bioprinting allows manufacturing three-dimensional (3D) structures that mimic tissues or organs. The bioinks used are mainly made of natural or synthetic polymers that must be biocompatible, printable, and biodegradable. These bioinks may incorporate progenitor cells, favoring graft implantation and regeneration of injured tissues. However, the natures of biomaterials, bioprinting processes, a lack of vascularization, and immune responses are factors that limit the viability and functionality of implanted cells and the regeneration of damaged tissues. These limitations can be addressed by incorporating extracellular vesicles (EV) into bioinks. Indeed, EV from progenitor cells may have regenerative capacities, being similar to those of their source cells. Therefore, their combinations with biomaterials can be used in cell-free therapies. Likewise, they can complement the manufacture of bioinks by increasing the viability, differentiation, and regenerative ability of incorporated cells. Thus, the main objective of this review is to show how the use of 3D bioprinting technology can be used for the application of EV in regenerative medicine by incorporating these nanovesicles into hydrogels used as bioinks. To this end, the latest advances derived from in vitro and in vivo studies have been described. Together, these studies show the high therapeutic potential of this strategy in regenerative medicine.
Collapse
Affiliation(s)
- Marta Camacho-Cardenosa
- Unidad de Gestión Clínica de Endocrinología y Nutrición-GC17, Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Hospital Universitario Reina Sofía, 14004 Córdoba, Spain; (M.C.-C.); (V.P.-E.); (G.E.-G.); (A.D.H.-M.)
| | - Victoria Pulido-Escribano
- Unidad de Gestión Clínica de Endocrinología y Nutrición-GC17, Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Hospital Universitario Reina Sofía, 14004 Córdoba, Spain; (M.C.-C.); (V.P.-E.); (G.E.-G.); (A.D.H.-M.)
| | - Guadalupe Estrella-Guisado
- Unidad de Gestión Clínica de Endocrinología y Nutrición-GC17, Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Hospital Universitario Reina Sofía, 14004 Córdoba, Spain; (M.C.-C.); (V.P.-E.); (G.E.-G.); (A.D.H.-M.)
| | - Gabriel Dorado
- Departamento Bioquímica y Biología Molecular, Campus Rabanales C6-1-E17, Campus de Excelencia Internacional Agroalimentario (ceiA3), Universidad de Córdoba, 14071 Córdoba, Spain;
- CIBER de Fragilidad y Envejecimiento Saludable (CIBERFES), 14004 Córdoba, Spain
| | - Aura D. Herrera-Martínez
- Unidad de Gestión Clínica de Endocrinología y Nutrición-GC17, Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Hospital Universitario Reina Sofía, 14004 Córdoba, Spain; (M.C.-C.); (V.P.-E.); (G.E.-G.); (A.D.H.-M.)
| | - María Ángeles Gálvez-Moreno
- Unidad de Gestión Clínica de Endocrinología y Nutrición-GC17, Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Hospital Universitario Reina Sofía, 14004 Córdoba, Spain; (M.C.-C.); (V.P.-E.); (G.E.-G.); (A.D.H.-M.)
| | - Antonio Casado-Díaz
- Unidad de Gestión Clínica de Endocrinología y Nutrición-GC17, Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Hospital Universitario Reina Sofía, 14004 Córdoba, Spain; (M.C.-C.); (V.P.-E.); (G.E.-G.); (A.D.H.-M.)
- CIBER de Fragilidad y Envejecimiento Saludable (CIBERFES), 14004 Córdoba, Spain
| |
Collapse
|
16
|
Garcia‐Aponte OF, Kahlenberg S, Kouroupis D, Egger D, Kasper C. Effects of Hydrogels on Mesenchymal Stem/Stromal Cells Paracrine Activity and Extracellular Vesicles Production. J Extracell Vesicles 2025; 14:e70057. [PMID: 40091440 PMCID: PMC11911545 DOI: 10.1002/jev2.70057] [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: 08/07/2024] [Revised: 12/10/2024] [Accepted: 02/11/2025] [Indexed: 03/19/2025] Open
Abstract
Mesenchymal stem/stromal cells (MSCs) are a valuable source of paracrine factors, as they have a remarkable secretory capacity, and there is a sizeable knowledge base to develop industrial and clinical production protocols. Promising cell-free approaches for tissue regeneration and immunomodulation are driving research towards secretome applications, among which extracellular vesicles (EVs) are steadily gaining attention. However, the manufacturing and application of EVs is limited by insufficient yields, knowledge gaps, and low standardization. Facing these limitations, hydrogels represent a versatile three-dimensional (3D) culture platform that can incorporate extracellular matrix (ECM) components to mimic the natural stem cell environment in vitro; via these niche-mimicking properties, hydrogels can regulate MSCs' morphology, adhesion, proliferation, differentiation and secretion capacities. However, the impact of the hydrogel's architectural, biochemical and biomechanical properties on the production of EVs remains poorly understood, as the field is still in its infancy and the interdependency of culture parameters compromises the comparability of the studies. Therefore, this review summarizes and discusses the reported effects of hydrogel encapsulation and culture on the secretion of MSC-EVs. Considering the effects of cell-material interactions on the overall paracrine activity of MSCs, we identify persistent challenges from low standardization and process control, and outline future paths of research, such as the synergic use of hydrogels and bioreactors to enhance MSC-EV generation.
Collapse
Affiliation(s)
- Oscar Fabian Garcia‐Aponte
- Department of Biotechnology and Food Science, Institute of Cell and Tissue Culture TechnologiesUniversity of Natural Resources and Life SciencesViennaAustria
| | - Simon Kahlenberg
- Department of Biotechnology and Food Science, Institute of Cell and Tissue Culture TechnologiesUniversity of Natural Resources and Life SciencesViennaAustria
| | - Dimitrios Kouroupis
- Department of Orthopedics, UHealth Sports Medicine Institute, Miller School of MedicineUniversity of MiamiMiamiFloridaUSA
- Diabetes Research Institute & Cell Transplant Center, Miller School of MedicineUniversity of MiamiMiamiFloridaUSA
| | - Dominik Egger
- Institute of Cell Biology and BiophysicsLeibniz University HannoverHannoverGermany
| | - Cornelia Kasper
- Department of Biotechnology and Food Science, Institute of Cell and Tissue Culture TechnologiesUniversity of Natural Resources and Life SciencesViennaAustria
| |
Collapse
|
17
|
Xiao N, Li Q, Liang G, Qian Z, Lin Y, Zhang H, Fu Y, Yang X, Zhang CT, Yang J, Liu A. Regulatory Roles of Exosomes in Aging and Aging-Related Diseases. Biogerontology 2025; 26:61. [PMID: 39966192 DOI: 10.1007/s10522-025-10200-7] [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: 11/09/2024] [Accepted: 01/29/2025] [Indexed: 02/20/2025]
Abstract
Exosomes are small vesicles with diameters ranging from 30 to 150 nm. They originate from cellular endocytic systems. These vesicles contain a rich payload of biomolecules, including proteins, nucleic acids, lipids, and metabolic products. Exosomes mediate intercellular communication and are key regulators of a diverse array of biological processes, such as oxidative stress and chronic inflammation. Furthermore, exosomes have been implicated in the pathogenesis of infectious diseases, autoimmune disorders, and cancer. Aging is closely associated with the onset and progression of numerous diseases and is significantly influenced by exosomes. Recent studies have consistently highlighted the important functions of exosomes in the regulation of cellular senescence. Additionally, research has explored their potential to delay aging, such as the alleviatory effects of stem cell-derived exosomes on the aging process, which offers broad potential for the development and application of exosomes as anti-aging therapeutic strategies. This review aims to comprehensively investigate the multifaceted impact of exosomes while concurrently evaluating their potential applications and underscoring their strategic significance in advancing anti-aging strategies.
Collapse
Affiliation(s)
- Nanyin Xiao
- Experimental Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, People's Republic of China
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Qiao Li
- Experimental Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, People's Republic of China
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Guangyu Liang
- Experimental Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, People's Republic of China
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Zonghao Qian
- Experimental Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, People's Republic of China
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Yan Lin
- Experimental Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, People's Republic of China
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
- Clinical Laboratory, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, People's Republic of China
| | - Heng Zhang
- Experimental Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, People's Republic of China
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Yangguang Fu
- Experimental Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, People's Republic of China
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Xiao Yang
- Experimental Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, People's Republic of China
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Cun-Tai Zhang
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Jiankun Yang
- Experimental Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, People's Republic of China
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
- Clinical Laboratory, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, People's Republic of China
| | - Anding Liu
- Experimental Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, People's Republic of China.
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China.
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China.
- Clinical Laboratory, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, People's Republic of China.
| |
Collapse
|
18
|
Chen Y, Qi W, Wang Z, Niu F. Exosome Source Matters: A Comprehensive Review from the Perspective of Diverse Cellular Origins. Pharmaceutics 2025; 17:147. [PMID: 40006514 PMCID: PMC11858990 DOI: 10.3390/pharmaceutics17020147] [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: 11/29/2024] [Revised: 01/02/2025] [Accepted: 01/14/2025] [Indexed: 02/27/2025] Open
Abstract
Exosomes have emerged as promising therapeutic agents in regenerative medicine. This review introduces a novel cell type-oriented perspective to systematically analyze exosomal properties in regenerative therapies. To our knowledge, this review is the first to comprehensively compare exosomes based on cellular source type, offering unprecedented insights into selecting optimal exosome producers for targeted regenerative applications. Factors beyond cellular origin influencing exosomal therapeutic efficacy, such as donor sites and collection methods, are also explored here. By synthesizing key advances, we propose promising research directions in the end. We aim to accelerate the development of more effective exosome-based regenerative therapies and highlight underexplored directions in this rapidly evolving field.
Collapse
Affiliation(s)
| | | | | | - Feng Niu
- Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 33 Badachu Road, Shijingshan, Beijing 100144, China; (Y.C.)
| |
Collapse
|
19
|
Gong Z, Chen Z, Li D, Lu X, Wu J, Sun H, Wang X, Liu S, Xia X, Lu F, Jiang J, Sun C, Wang H, Zeng F, Ma X. Hydrogel loaded with cerium-manganese nanoparticles and nerve growth factor enhances spinal cord injury repair by modulating immune microenvironment and promoting neuronal regeneration. J Nanobiotechnology 2025; 23:29. [PMID: 39833803 PMCID: PMC11748312 DOI: 10.1186/s12951-025-03098-3] [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: 09/08/2024] [Accepted: 01/07/2025] [Indexed: 01/22/2025] Open
Abstract
BACKGROUND Spinal cord injury (SCI) treatment remains a formidable challenge, as current therapeutic approaches provide only marginal relief and fail to reverse the underlying tissue damage. This study aims to develop a novel composite material combining enzymatic nanoparticles and nerve growth factor (NGF) to modulate the immune microenvironment and enhance SCI repair. METHODS CeMn nanoparticles (NP) and CeMn NP-polyethylene glycol (PEG) nanozymes were synthesized via sol-gel reaction and DSPE-mPEG modification. Transmission Electron Microscopy, Selected-Area Electron Diffraction, X-ray Diffraction and X-ray Photoelectron Spectroscopy confirmed their crystalline structure, mixed-valence states, and redox properties. Size uniformity, biocompatibility, and catalytic activity were assessed via hydrodynamic diameter, zeta potential, and elemental analysis. The Lightgel/NGF/CeMn NP-PEG composite was synthesized and characterized via electron microscopy, compression testing, rheological analysis, NGF release kinetics, and 30-day degradation studies. Both in vitro and in vivo experiments were conducted to evaluate the therapeutic effects of the composite on SCI. RESULTS The Lightgel/NGF/CeMn NP-PEG composite was successfully synthesized, exhibiting favorable physical properties. At a CeMn NP-PEG concentration of 4 µg/mL, the composite maintained cell viability and demonstrated enhanced biological activity. It also showed superior mechanical properties and an effective NGF release profile. Notably, the composite significantly upregulated the expression of nerve growth-associated proteins, reduced inflammatory cytokines, scavenged reactive oxygen species (ROS), and promoted M2 macrophage polarization by inhibiting the cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) signaling pathway. In a rat SCI model, it facilitated functional recovery and attenuated inflammation. CONCLUSION The Lightgel/NGF/CeMn NP-PEG composite shows significant therapeutic promise for SCI, effectively eliminating ROS, promoting M2 macrophage polarization, reducing pro-inflammatory cytokines, and supporting neuronal regeneration. These effects substantially enhance motor function in SCI rats, positioning it as a promising candidate for future clinical applications.
Collapse
Affiliation(s)
- Zhaoyang Gong
- Department of Orthopedics, Huashan Hospital, Fudan University, No. 12, Middle Wulumuqi Road, Jing'an District, Shanghai, 200040, China
| | - Zhenhao Chen
- Department of Orthopedics, Huashan Hospital, Fudan University, No. 12, Middle Wulumuqi Road, Jing'an District, Shanghai, 200040, China
| | - Dachuan Li
- Department of Orthopedics, Huashan Hospital, Fudan University, No. 12, Middle Wulumuqi Road, Jing'an District, Shanghai, 200040, China
| | - Xiao Lu
- Department of Orthopedics, Huashan Hospital, Fudan University, No. 12, Middle Wulumuqi Road, Jing'an District, Shanghai, 200040, China
| | - Jianwei Wu
- Department of Orthopedics, Huashan Hospital, Fudan University, No. 12, Middle Wulumuqi Road, Jing'an District, Shanghai, 200040, China
| | - Hanqiu Sun
- Department of Orthopedics, Huashan Hospital, Fudan University, No. 12, Middle Wulumuqi Road, Jing'an District, Shanghai, 200040, China
| | - Ximeng Wang
- Department of Orthopedics, Huashan Hospital, Fudan University, No. 12, Middle Wulumuqi Road, Jing'an District, Shanghai, 200040, China
| | - Siyang Liu
- Department of Orthopedics, Huashan Hospital, Fudan University, No. 12, Middle Wulumuqi Road, Jing'an District, Shanghai, 200040, China
| | - Xinlei Xia
- Department of Orthopedics, Huashan Hospital, Fudan University, No. 12, Middle Wulumuqi Road, Jing'an District, Shanghai, 200040, China
| | - Feizhou Lu
- Department of Orthopedics, Huashan Hospital, Fudan University, No. 12, Middle Wulumuqi Road, Jing'an District, Shanghai, 200040, China
| | - Jianyuan Jiang
- Department of Orthopedics, Huashan Hospital, Fudan University, No. 12, Middle Wulumuqi Road, Jing'an District, Shanghai, 200040, China
| | - Chi Sun
- Department of Orthopedics, Huashan Hospital, Fudan University, No. 12, Middle Wulumuqi Road, Jing'an District, Shanghai, 200040, China.
| | - Hongli Wang
- Department of Orthopedics, Huashan Hospital, Fudan University, No. 12, Middle Wulumuqi Road, Jing'an District, Shanghai, 200040, China.
| | - Feng Zeng
- Artemisinin Research Center, Institute of Science and Technology, The First Affiliated Hospital, The First Clinical Medical School, Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510450, China.
| | - Xiaosheng Ma
- Department of Orthopedics, Huashan Hospital, Fudan University, No. 12, Middle Wulumuqi Road, Jing'an District, Shanghai, 200040, China.
| |
Collapse
|
20
|
Guo J, Huang Z, Wang Q, Wang M, Ming Y, Chen W, Huang Y, Tang Z, Huang M, Liu H, Jia B. Opportunities and challenges of bacterial extracellular vesicles in regenerative medicine. J Nanobiotechnology 2025; 23:4. [PMID: 39754127 PMCID: PMC11697683 DOI: 10.1186/s12951-024-02935-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 10/16/2024] [Indexed: 01/07/2025] Open
Abstract
Extracellular vesicles (EVs) are membrane-bound vesicles that are shed or secreted from the cell membrane and enveloped by a lipid bilayer. They possess stability, low immunogenicity, and non-cytotoxicity, exhibiting extensive prospects in regenerative medicine (RM). However, natural EVs pose challenges, such as insufficient targeting capabilities, potential biosafety concerns, and limited acquisition pathways. Although engineered EVs demonstrate excellent therapeutic efficacy, challenges such as low production yield and the complexity of engineering modifications constrain their further clinical applications. Bacteria have advantages such as rapid proliferation, diverse gene editing methods, mature cultivation techniques, and relatively easy preparation of bacterial EVs (BEVs), which can be used to effectively address the challenges currently encountered in the field of EVs. This review provides a description of the biogenesis and pathophysiological functions of BEVs, and strategies for optimizing BEVs preparation to attain efficiency and safety are discussed. An analysis of natural characteristics of BEVs is also conducted to explore how to leverage their advantages or mitigate their limitations, thereby overcoming constraints on the application of BEVs in RM. In summary, engineered BEVs possess characteristics such as high production yield, excellent stability, and high drug-delivering capabilities, laying the foundation for their application in RM.
Collapse
Affiliation(s)
- Jiming Guo
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China
| | - Zhijie Huang
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China
| | - Qinjing Wang
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China
| | - Min Wang
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China
| | - Yue Ming
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China
| | - Weixing Chen
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China
| | - Yisheng Huang
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China
| | - Zhengming Tang
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China
| | - Mingshu Huang
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China
| | - Hongyu Liu
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China
| | - Bo Jia
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
21
|
Zeng X, Liao Y, Huang D, Yang J, Dai Z, Chen Z, Luo X, Gong H, Huang S, Zhang L. Exosomes derived from hUC-MSCs exhibit ameliorative efficacy upon previous cesarean scar defect via orchestrating β-TrCP/CHK1 axis. Sci Rep 2025; 15:489. [PMID: 39747551 PMCID: PMC11697314 DOI: 10.1038/s41598-024-84689-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Accepted: 12/26/2024] [Indexed: 01/04/2025] Open
Abstract
Previous cesarean scar defect (PCSD), also acknowledged as the myometrium of uterus defects, which commonly results in myometrial discontinuity between the uterine and cervical cavity. Current literatures have indicated the efficacy of MSCs and MSC-derived exosomes (MSC-Exos) for diverse refractory disease administration, yet the feasibility of MSC-Exos for PCSD treatment is largely obscure. In this study, we took advantage of the in vivo myofibrotic model for mimicking the typical manifestation of PCSD and the assessment of fertility. Meanwhile, the ex vivo scratch wound healing model is used for exploring the underlying molecular mechanism. On the one hand, we took advantage of the TGF-β-induced in vitro myofibrotic model and the full-thickness uterine injury rat model to verify the efficacy of human umbilical cord MSC-derived exosomes (hUC-MSC-Exos). On the other hand, with the aid of CHK1 overexpression and β-TrCP knockdown, together with multifaceted biological analyses (e.g., histopathological sections, qRT-PCR assay, western-blotting analysis, Co-IP assay, protein degradation and ubiquitination), we further dissected the underlying regulatory mechanism. We identified hUC-MSC-Exos and verified the suppressive effect of hUC-MSC-Exos upon TGF-β-induced in vitro myofibrotic model and full-thickness uterine injury in rats via delivering β-TrCP. Furthermore, we found that β-TrCP in hUC-MSC-Exos facilitated the ubiquitination degradation of CHK1 and inhibited the myofibrosis. Collectively, our data indicated the preferable ameliorative effect of hUC-MSC-Exos upon both the in vitro and in vivo myofibrotic models, together with the β-TrCP and CHK1-mediated regulatory mechanism. Our findings provided new references of hUC-MSC-Exos-based regimens for PCSD management in future.
Collapse
Affiliation(s)
- Xiaoling Zeng
- Department of Obstetrics, Guizhou Provincial People's Hospital, Guiyang, 550001, Guizhou, China
| | - Yuan Liao
- Department of Obstetrics, Guizhou Provincial People's Hospital, Guiyang, 550001, Guizhou, China
| | - Dan Huang
- Department of Obstetrics, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Jing Yang
- Department of Obstetrics, Guizhou Provincial People's Hospital, Guiyang, 550001, Guizhou, China
| | - Zhihua Dai
- Stem Cell Bank of Guizhou Province, Guizhou Health-Biotech Biotechnology Co., Ltd., Guiyang, 550000, China
| | - Zhengyong Chen
- Department of Obstetrics, Guizhou Provincial People's Hospital, Guiyang, 550001, Guizhou, China
| | - Xin Luo
- The First Affiliated Hospital of Jinan University, Guangzhou, 510632, Guangdong, China
| | - Han Gong
- Department of Obstetrics, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Shengwen Huang
- Prenatal Diagnosis Center, Guizhou Provincial People's Hospital, Guiyang, 550001, Guizhou, China.
| | - Leisheng Zhang
- Science and Technology Innovation Center, Shandong Provincial Key Medical and Health Laboratory of Blood Ecology and Biointelligence, Jinan Key Laboratory of Medical Cell Bioengineering, Cardio- cerebrovascular Disease Hospital of Jinan, The Fourth People's Hospital of Jinan, The Teaching Hospital of Shandong Second Medical University, 50 Shifan Road, Tianqiao District, Jinan, 250031, Shandong, China.
- National Health Commission (NHC) Key Laboratory of Diagnosis and Therapy of Gastrointestinal Tumor, Gansu Provincial Hospital, The Third Hospital of Lanzhou University (The Third Clinical College), Lanzhou, 730000, China.
| |
Collapse
|
22
|
Thai NLB, Fittante E, Ma Z, Monroe MB. Rapid Fabrication of Polyvinyl Alcohol Hydrogel Foams With Encapsulated Mesenchymal Stem Cells for Chronic Wound Treatment. J Biomed Mater Res A 2025; 113:e37868. [PMID: 39794931 DOI: 10.1002/jbm.a.37868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 12/17/2024] [Accepted: 12/26/2024] [Indexed: 01/13/2025]
Abstract
Chronic wounds present a major healthcare challenge around the world, and significant hurdles remain in their effective treatment due to limitations in accessible treatment options. Mesenchymal stem cells (MSCs) with multifunctional differentiation and modulatory properties have been delivered to chronic wounds to enhance closure but have limited engraftment when delivered without a scaffold. In this study, hybrid porous hydrogel foams composed of modified polyvinyl alcohol and gelatin were developed that are suitable for rapid and facile MSC encapsulation, fully degradable, and supportive of wound healing. Rapid fabrication and encapsulation within porous foams was achieved using a cytocompatible gas blowing process. The hybrid hydrogels have tunable degradation rates based on chemistry, with complete mass loss achieved within 2-6 weeks, which is compatible with chronic wound closure rates. High encapsulated A375 epithelial cell and MSC viability with maintained cell functionality over 2 weeks reveals the potential of these hydrogels to serve as cell delivery systems for chronic wound treatment. An ex vivo porcine skin wound model demonstrated enhanced healing after application of cell-laden hydrogel foams. Overall, hybrid hydrogel foams with encapsulated therapeutic cells have the capacity for robust wound healing and are a promising platform for chronic wound dressings.
Collapse
Affiliation(s)
- Nghia Le Ba Thai
- Biomedical and Chemical Engineering and BioInspired Syracuse: Institute for Material and Living Systems, Syracuse University, Syracuse, New York, USA
| | - Emily Fittante
- Biomedical and Chemical Engineering and BioInspired Syracuse: Institute for Material and Living Systems, Syracuse University, Syracuse, New York, USA
| | - Zhen Ma
- Biomedical and Chemical Engineering and BioInspired Syracuse: Institute for Material and Living Systems, Syracuse University, Syracuse, New York, USA
| | - Mary Beth Monroe
- Biomedical and Chemical Engineering and BioInspired Syracuse: Institute for Material and Living Systems, Syracuse University, Syracuse, New York, USA
| |
Collapse
|
23
|
Qi L, Hu L, Qian R, Ye B, Feng Y, Deng Y, Wang C, Zhou C, Liu G, Gao X, Lin C, Ding Q, Song C, Zhao Z, Lin Z, Zhu J, Zhang M. Advances in mesenchymal stem cell-centered stem cell therapy in the treatment of hypoxic-ischemic injury. Int Immunopharmacol 2024; 143:113430. [PMID: 39437489 DOI: 10.1016/j.intimp.2024.113430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 10/03/2024] [Accepted: 10/15/2024] [Indexed: 10/25/2024]
Abstract
Hypoxic-ischemic brain damage (HIBD) is a leading cause of neonatal death and neurological dysfunction for which no particularly effective treatment is available. Stem cells possess multi-directional differentiation potential and can secrete a variety of cytokines. They not only have the ability to replace tissue and repair lesions but also improve neurological damage caused by HIBD through paracrine mechanisms, including anti-apoptosis, reduction of inflammation, and promotion of endogenous repair. Recently, as research on stem cells, particularly mesenchymal stem cells, has deepened, the application of stem cells in treating HIBD has become a prominent research topic, yielding fruitful results, particularly regarding the neuroprotective effects and mechanisms of the stem cell paracrine pathway. With advances in stem cell injection, distribution, and biomaterial incorporation, applications of stem cells have become more widespread and comprehensive. This review summarizes and discusses the research progress on stem cells in HIBD treatment to provide theoretical support for HIBD treatment and enhance the feasibility of clinical translation.
Collapse
Affiliation(s)
- Lixin Qi
- Department of Neonatology, the Second School of Medicine, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Lei Hu
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Rengcheng Qian
- Department of Neonatology, the Second School of Medicine, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Perinatal Medicine of Wenzhou, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Structural Malformations in Children of Zhejiang Province, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Zhejiang Provincial Clinical Research Center for Pediatric Disease, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Bangming Ye
- Department of Neonatology, the Second School of Medicine, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Perinatal Medicine of Wenzhou, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Structural Malformations in Children of Zhejiang Province, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Zhejiang Provincial Clinical Research Center for Pediatric Disease, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yani Feng
- Department of Neonatology, the Second School of Medicine, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Perinatal Medicine of Wenzhou, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Structural Malformations in Children of Zhejiang Province, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Zhejiang Provincial Clinical Research Center for Pediatric Disease, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yixuan Deng
- School of Ophthalmology & Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Chenyi Wang
- The First School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Chunting Zhou
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Guanhao Liu
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xiuying Gao
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Congying Lin
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Qiang Ding
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Chunyu Song
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Ziming Zhao
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zhenlang Lin
- Department of Neonatology, the Second School of Medicine, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Perinatal Medicine of Wenzhou, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Structural Malformations in Children of Zhejiang Province, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Zhejiang Provincial Clinical Research Center for Pediatric Disease, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.
| | - Jianghu Zhu
- Department of Neonatology, the Second School of Medicine, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Perinatal Medicine of Wenzhou, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Structural Malformations in Children of Zhejiang Province, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Zhejiang Provincial Clinical Research Center for Pediatric Disease, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.
| | - Min Zhang
- Department of Neonatology, the Second School of Medicine, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Perinatal Medicine of Wenzhou, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Structural Malformations in Children of Zhejiang Province, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Zhejiang Provincial Clinical Research Center for Pediatric Disease, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.
| |
Collapse
|
24
|
Li B, Chen H, Hang R. Osseointegration-Related Exosomes for Surface Functionalization of Titanium Implants. Biomater Res 2024; 28:0124. [PMID: 39711824 PMCID: PMC11661649 DOI: 10.34133/bmr.0124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 11/05/2024] [Accepted: 11/27/2024] [Indexed: 12/24/2024] Open
Abstract
Despite that the clinical application of titanium-based implants has achieved great success, patients' own diseases and/or unhealthy lifestyle habits often lead to implant failure. Many studies have been carried out to modify titanium implants to promote osseointegration and implant success. Recent studies showed that exosomes, proactively secreted extracellular vesicles by mammalian cells, could selectively target and modulate the functions of recipient cells such as macrophages, nerve cells, endothelial cells, and bone marrow mesenchymal stem cells that are closely involved in implant osseointegration. Accordingly, using exosomes to functionalize titanium implants has been deemed as a novel and effective way to improve their osseointegration ability. Herein, recent advances pertaining to surface functionalization of titanium implants with exosomes are analyzed and discussed, with focus on the role of exosomes in regulating the functions of osseointegration-related cells, and their immobilization strategies as well as resultant impact on osseointegration ability.
Collapse
Affiliation(s)
- Boqiong Li
- Department of Materials Science and Engineering,
Jinzhong University, Jinzhong 030619, China
| | - Huanming Chen
- Shanxi Key Laboratory of Biomedical Metal Materials, College of Materials Science and Engineering,
Taiyuan University of Technology, Taiyuan 030024, China
| | - Ruiqiang Hang
- Shanxi Key Laboratory of Biomedical Metal Materials, College of Materials Science and Engineering,
Taiyuan University of Technology, Taiyuan 030024, China
| |
Collapse
|
25
|
Khosroshahi PA, Ghanbari M. MicroRNA dysregulation in glutamate and dopamine pathways of schizophrenia: From molecular pathways to diagnostic and therapeutic approaches. Prog Neuropsychopharmacol Biol Psychiatry 2024; 135:111081. [PMID: 39002925 DOI: 10.1016/j.pnpbp.2024.111081] [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: 12/01/2023] [Revised: 06/28/2024] [Accepted: 07/07/2024] [Indexed: 07/15/2024]
Abstract
Schizophrenia is a complex psychiatric disorder, and genetic and environmental factors have been implicated in its development. Dysregulated glutamatergic and dopaminergic transmission pathways are involved in schizophrenia development. Besides genetic mutations, epigenetic dysregulation has a considerable role in dysregulating molecular pathways involved in schizophrenia. MicroRNAs (miRNAs) are small, non-coding RNAs that target specific mRNAs and inhibit their translation into proteins. As epigenetic factors, miRNAs regulate many genes involved in glutamate and dopamine signaling pathways; thereby, their dysregulation can contribute to the development of schizophrenia. Secretion of specific miRNAs from damaged cells into body fluids can make them one of the ideal non-invasive biomarkers in the early diagnosis of schizophrenia. Also, understanding the molecular mechanisms of miRNAs in schizophrenia pathogenesis can pave the way for developing novel treatments for patients with schizophrenia. In this study, we reviewed the glutamatergic and dopaminergic pathophysiology and highlighted the role of miRNA dysregulation in schizophrenia development. Besides, we shed light on the significance of circulating miRNAs for schizophrenia diagnosis and the recent findings on the miRNA-based treatment for schizophrenia.
Collapse
Affiliation(s)
| | - Mohammad Ghanbari
- Department of Animal Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran.
| |
Collapse
|
26
|
Teymouri S, Yousefi MH, Heidari S, Farokhi S, Afkhami H, Kashfi M. Beyond antibiotics: mesenchymal stem cells and bacteriophages-new approaches to combat bacterial resistance in wound infections. Mol Biol Rep 2024; 52:64. [PMID: 39699690 DOI: 10.1007/s11033-024-10163-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Accepted: 12/09/2024] [Indexed: 12/20/2024]
Abstract
Wound management is a major global health problem. With the rising incidence of diabetic wounds, accidents, and other injuries, the demand for prompt wound treatment has become increasingly critical. Millions of people suffer from serious, large wounds resulting from major accidents, surgeries, and wars. These wounds require considerable time to heal and are susceptible to infection. Furthermore, chronic wounds, particularly in elderly and diabetic patients, often require frequent medical interventions to prevent complications. Consequently, wound management imposes a significant economic burden worldwide. The complications arising from wound infections can vary from localized issues to systemic effects. The most severe local complication of wound infection is the non-healing, which results from the disruption of the wound-healing process. This often leads to significant pain, discomfort, and psychological trauma for the patient. Systemic complications may include cellulitis, osteomyelitis, and septicemia. Mesenchymal stem cells are characterized by their high capacity for division, making them suitable candidates for the treatment of tissue damage. Additionally, they produce antimicrobial peptides and various cytokines, which enhance their antimicrobial activity. Evidence shows that phages are effective in treating wound-related infections, and phage therapy has proven to be highly effective for patients when administered correctly. The purpose of this article is to explore the use of bacteriophages and mesenchymal stem cells in wound healing and infection management.
Collapse
Affiliation(s)
- Samane Teymouri
- Department of Microbiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Hasan Yousefi
- Department of Tissue Engineering and Applied Cell Sciences, School of Medicine, Qom University of Medical Sciences, Qom, Iran
- Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran
| | | | - Simin Farokhi
- Student Research Committee, USERN Office, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Hamed Afkhami
- Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran.
- Nervous System Stem Cells Research Center, Semnan University of Medical Sciences, Semnan, Iran.
- Department of Medical Microbiology, Faculty of Medicine, Shahed University, Tehran, Iran.
| | - Mojtaba Kashfi
- Nervous System Stem Cells Research Center, Semnan University of Medical Sciences, Semnan, Iran.
- Fellowship in Clinical Laboratory Sciences, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
27
|
Lin X, Fang Y, Mi X, Fu J, Chen S, Wu M, Jin N. Intrauterine injection of bioengineered hydrogel loaded exosomes derived from HUCM stem cells and spermidine prominently augments the pregnancy rate in thin endometrium rats. Regen Ther 2024; 27:63-72. [PMID: 38525237 PMCID: PMC10959642 DOI: 10.1016/j.reth.2024.02.003] [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: 11/30/2023] [Revised: 01/10/2024] [Accepted: 02/16/2024] [Indexed: 03/26/2024] Open
Abstract
The endometrium is essential to the development of embryos and pregnancy. Human umbilical cord mesenchymal stem cells (HUCMSCs) are promising stem cell sources. HUCMSCs self-renew quickly and are painless to collect. Spermidine is an inherent polyamine needed for cellular and molecular processes that regulate physiology and function. HUCMSCs and spermidine (SN) may heal intrauterine adhesions. HUCMSCs were investigated for endometrial repair in rats. Composite hydrogels are used for medical exosome implantation, including their materials, properties, and embedding procedures. This study examined whether bioengineered hydrogel-loaded exosomes from HUCMSCs and spermidine prenatally improved conception rates in mice with poor endometrial lining. The data show that HUCMSC and SN provide a good experimental base for HUCMSC safety and intrauterine treatment in rats. Western blots, exosome structural analysis, pregnancy outcomes, flow cytometry, H&E staining, immunohistochemistry, and immunofluorescence labelling found and recovered the aberrant area. HUCM-derived stem cells and spermidine-derived exosomes biophysically match. These traits strengthen and prolong endometrial function. Pregnant rats with HUCMSC and SN had thinner endometrium. Hydrogel-incorporated HEHUCMSC and SN exosomes may improve IUI in rats with thin endometrium.
Collapse
Affiliation(s)
- Xiuying Lin
- Department of Pathology and Pathophysiology, School of Basic Medical Sciences, Yanbian University, Yanbian 133002, China
- Center of Reproductive Medicine, Jilin Province People's Hospital, Changchun 130021, China
| | - Yanqiu Fang
- Center of Reproductive Medicine, Jilin Province People's Hospital, Changchun 130021, China
| | - Xuguang Mi
- Center of Reproductive Medicine, Jilin Province People's Hospital, Changchun 130021, China
| | - Jianhua Fu
- Center of Reproductive Medicine, Jilin Province People's Hospital, Changchun 130021, China
| | - Shiling Chen
- Center of Reproductive Medicine, Jilin Province People's Hospital, Changchun 130021, China
| | - Mengxue Wu
- Center of Reproductive Medicine, Jilin Province People's Hospital, Changchun 130021, China
| | - Ningyi Jin
- Department of Pathology and Pathophysiology, School of Basic Medical Sciences, Yanbian University, Yanbian 133002, China
- Changchun Institute of Veterinary Medicine, Chinese Academy of Agricultural Sciences, Changchun 130000, China
| |
Collapse
|
28
|
Aliniay-Sharafshadehi S, Yousefi MH, Ghodratie M, Kashfi M, Afkhami H, Ghoreyshiamiri SM. Exploring the therapeutic potential of different sources of mesenchymal stem cells: a novel approach to combat burn wound infections. Front Microbiol 2024; 15:1495011. [PMID: 39678916 PMCID: PMC11638218 DOI: 10.3389/fmicb.2024.1495011] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 11/08/2024] [Indexed: 12/17/2024] Open
Abstract
The most prevalent and harmful injuries are burns, which are still a major global health problem. Burn injuries can cause issues because they boost the inflammatory and metabolic response, which can cause organ malfunction and systemic failure. On the other hand, a burn wound infection creates an environment that is conducive to the growth of bacteria and might put the patient at risk for sepsis. In addition, scarring is unavoidable, and this results in patients having functional and cosmetic issues. Wound healing is an amazing phenomenon with a complex mechanism that deals with different types of cells and biomolecules. Cell therapy using stem cells is one of the most challenging treatment methods that accelerates the healing of burn wounds. Since 2000, the use of mesenchymal stem cells (MSCs) in regenerative medicine and wound healing has increased. They can be extracted from various tissues, such as bone marrow, fat, the umbilical cord, and the amniotic membrane. According to studies, stem cell therapy for burn wounds increases angiogenesis, has anti-inflammatory properties, slows the progression of fibrosis, and has an excellent ability to differentiate and regenerate damaged tissue. Figuring out the main preclinical and clinical problems that stop people from using MSCs and then suggesting the right ways to improve therapy could help show the benefits of MSCs and move stem cell-based therapy forward. This review's objective was to assess mesenchymal stem cell therapy's contribution to the promotion of burn wound healing.
Collapse
Affiliation(s)
- Shahrzad Aliniay-Sharafshadehi
- Department of Microbiology, Faculty of Advanced Science and Technology, Tehran Medical Science, Islamic Azad University, Tehran, Iran
| | - Mohammad Hasan Yousefi
- Department of Tissue Engineering and Applied Cell Sciences, School of Medicine, Qom University of Medical Sciences, Qom, Iran
- Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran
| | - Mohammad Ghodratie
- Department of Medical Microbiology, Faculty of Medicine, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Mojtaba Kashfi
- Fellowship in Clinical Laboratory Sciences, Mashhad University of Medical Sciences, Mashhad, Iran
- Nervous System Stem Cells Research Center, Semnan University of Medical Sciences, Semnan, Iran
| | - Hamed Afkhami
- Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran
- Nervous System Stem Cells Research Center, Semnan University of Medical Sciences, Semnan, Iran
- Department of Medical Microbiology, Faculty of Medicine, Shahed University, Tehran, Iran
| | | |
Collapse
|
29
|
Assi MM, Grawish ME, Elsabaa HM, Helal ME, Ezzat SK. Therapeutic potential of hyaluronic acid hydrogel combined with bone marrow stem cells-conditioned medium on arthritic rats' TMJs. Sci Rep 2024; 14:26828. [PMID: 39500985 PMCID: PMC11538243 DOI: 10.1038/s41598-024-77325-6] [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: 05/02/2024] [Accepted: 10/21/2024] [Indexed: 11/08/2024] Open
Abstract
Conditioned media (CM) is derived from mesenchymal stem cells (MSC) culture and contains biologically active components. CM is easy to handle and reduces inflammation while repairing injured joints. Combination therapy of the CM with cross-linked hyaluronic acid (HA) could ameliorate the beneficial effect of HA in treating degenerative changes of articulating surfaces associated with arthritic rats' temporomandibular joints (TMJs). This study aimed to evaluate the therapeutic potential of HA hydrogel combined with bone marrow stem cells-conditioned medium (BMSCs-CM) on the articulating surfaces of TMJs associated with complete Freund's adjuvant (CFA)-induced arthritis. Fifty female Sprague-Dawley rats were divided randomly into five equal groups. Rats of group I served as the negative controls and received intra-articular (IA) injections of 50 µl saline solution, whereas rats of group II were subjected to twice IA injections of 50 µg CFA in 50 µl; on day 1 of the experiment to induce persistent inflammation and on day 14 to induce arthritis. Rats of group III and IV were handled as group II and instead, they received an IA injection of 50 µl HA hydrogel and 50 µl of BMSCs-CM, respectively. Rats of group V were given combined IA injections of 50 µl HA hydrogel and BMSCs-CM. All rats were euthanized after the 4th week of inducing arthritis. The joints were processed for sectioning and histological staining using hematoxylin and eosin, Masson's trichrome and toluidine blue special staining, and immunohistochemical staining for nuclear factor-kappa B (NF-κB). SPSS software was used to analyze the data and one-way analysis of variance followed by post-hoc Tukey statistical tests were used to test the statistical significance at 0.05 for alpha and 0.2 for beta. In the pooled BMSC-CM, 197.14 pg/ml of platelet-derived growth factor and 112.22 pg/ml of interleukin-10 were detected. Compared to TMJs of groups III and IV, TMJs of group V showed significant improvements (P = 0.001) in all parameters tested as the disc thickness was decreased (331.79 ± 0.73), the fibrocartilaginous layer was broadened (0.96 ± 0.04), and the amount of the trabecular bone was distinctive (19.35 ± 1.07). The mean values for the collagen amount were increased (12.29 ± 1.38) whereas the mean values for the NF-κB expression were decreased (0.62 ± 0.15). Combination therapy of HA hydrogel and BMSCs-CM is better than using HA hydrogel or BMSCs-CM, separately to repair degenerative changes in rats' TMJs associated with CFA-induced arthritis.
Collapse
Affiliation(s)
- Mai M Assi
- Department of Oral Biology, Faculty of Dentistry, Mansoura University, Mansoura, 35511, Egypt
| | - Mohammed E Grawish
- Department of Oral Biology, Faculty of Dentistry, Mansoura University, Mansoura, 35511, Egypt.
- Department of Oral Biology, Faculty of Oral and Dental Medicine, Delta University for Science and Technology, Dakahlia, Egypt.
| | - Heba Mahmoud Elsabaa
- Department of Oral Biology, Faculty of Dentistry, Mansoura University, Mansoura, 35511, Egypt
- Department of Oral Biology and Pathology, Faculty of Dentistry, Badr University in Cairo, Cairo, Egypt
| | - Mohamad E Helal
- Department of Oral Biology, Faculty of Dentistry, Mansoura University, Mansoura, 35511, Egypt
| | - Samah K Ezzat
- Department of Oral Biology, Faculty of Dentistry, Mansoura University, Mansoura, 35511, Egypt
| |
Collapse
|
30
|
Lu Y, Shi R, He W, An Q, Zhao J, Gao X, Zhang B, Zhang L, Xu K, Ma D. Cell therapy in Sjögren's syndrome: opportunities and challenges. Expert Rev Mol Med 2024; 26:e28. [PMID: 39438246 PMCID: PMC11505611 DOI: 10.1017/erm.2024.21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 06/26/2024] [Accepted: 07/09/2024] [Indexed: 10/25/2024]
Abstract
Sjögren's syndrome (SS) is a chronic autoimmune disease caused by immune system disorders. The main clinical manifestations of SS are dry mouth and eyes caused by the destruction of exocrine glands, such as the salivary and lacrimal glands, and systemic manifestations, such as interstitial pneumonia, interstitial nephritis and vasculitis. The pathogenesis of this condition is complex. However, this has not been fully elucidated. Treatment mainly consists of glucocorticoids, disease-modifying antirheumatic drugs and biological agents, which can only control inflammation but not repair the tissue. Therefore, identifying methods to regulate immune disorders and repair damaged tissues is imperative. Cell therapy involves the transplantation of autologous or allogeneic normal or bioengineered cells into the body of a patient to replace damaged cells or achieve a stronger immunomodulatory capacity to cure diseases, mainly including stem cell therapy and immune cell therapy. Cell therapy can reduce inflammation, relieve symptoms and promote tissue repair and regeneration of exocrine glands such as the salivary glands. It has broad application prospects and may become a new treatment strategy for patients with SS. However, there are various challenges in cell preparation, culture, storage and transportation. This article reviews the research status and prospects of cell therapies for SS.
Collapse
Affiliation(s)
- Yangyang Lu
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
- Shanxi Province Clinical Research Center for Dermatologic and Immunologic Diseases (Rheumatic diseases), Taiyuan, China
- Shanxi Province Clinical Theranostics Technology Innovation Center for Immunologic and Rheumatic Diseases, Taiyuan, China
| | - Rongjing Shi
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
- Shanxi Province Clinical Research Center for Dermatologic and Immunologic Diseases (Rheumatic diseases), Taiyuan, China
- Shanxi Province Clinical Theranostics Technology Innovation Center for Immunologic and Rheumatic Diseases, Taiyuan, China
| | - Wenqin He
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
- Shanxi Province Clinical Research Center for Dermatologic and Immunologic Diseases (Rheumatic diseases), Taiyuan, China
- Shanxi Province Clinical Theranostics Technology Innovation Center for Immunologic and Rheumatic Diseases, Taiyuan, China
| | - Qi An
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
- Shanxi Province Clinical Research Center for Dermatologic and Immunologic Diseases (Rheumatic diseases), Taiyuan, China
- Shanxi Province Clinical Theranostics Technology Innovation Center for Immunologic and Rheumatic Diseases, Taiyuan, China
| | - Jingwen Zhao
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
- Shanxi Province Clinical Research Center for Dermatologic and Immunologic Diseases (Rheumatic diseases), Taiyuan, China
- Shanxi Province Clinical Theranostics Technology Innovation Center for Immunologic and Rheumatic Diseases, Taiyuan, China
| | - Xinnan Gao
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
- Shanxi Province Clinical Research Center for Dermatologic and Immunologic Diseases (Rheumatic diseases), Taiyuan, China
- Shanxi Province Clinical Theranostics Technology Innovation Center for Immunologic and Rheumatic Diseases, Taiyuan, China
| | - Baiyan Zhang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
- Shanxi Province Clinical Research Center for Dermatologic and Immunologic Diseases (Rheumatic diseases), Taiyuan, China
- Shanxi Province Clinical Theranostics Technology Innovation Center for Immunologic and Rheumatic Diseases, Taiyuan, China
| | - Liyun Zhang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
- Shanxi Province Clinical Research Center for Dermatologic and Immunologic Diseases (Rheumatic diseases), Taiyuan, China
- Shanxi Province Clinical Theranostics Technology Innovation Center for Immunologic and Rheumatic Diseases, Taiyuan, China
| | - Ke Xu
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
- Shanxi Province Clinical Research Center for Dermatologic and Immunologic Diseases (Rheumatic diseases), Taiyuan, China
- Shanxi Province Clinical Theranostics Technology Innovation Center for Immunologic and Rheumatic Diseases, Taiyuan, China
| | - Dan Ma
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
- Shanxi Province Clinical Research Center for Dermatologic and Immunologic Diseases (Rheumatic diseases), Taiyuan, China
- Shanxi Province Clinical Theranostics Technology Innovation Center for Immunologic and Rheumatic Diseases, Taiyuan, China
- Shanxi Academy of Advanced Research and Innovation, Taiyuan, China
| |
Collapse
|
31
|
Nikolits I, Chariyev-Prinz F, Egger D, Liebner F, Mytzka N, Kasper C. Characterization of MSC Growth, Differentiation, and EV Production in CNF Hydrogels Under Static and Dynamic Cultures in Hypoxic and Normoxic Conditions. Bioengineering (Basel) 2024; 11:1050. [PMID: 39451425 PMCID: PMC11504186 DOI: 10.3390/bioengineering11101050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 10/14/2024] [Accepted: 10/20/2024] [Indexed: 10/26/2024] Open
Abstract
Mesenchymal stem cells (MSCs) hold immense therapeutic potential due to their regenerative and immunomodulatory properties. However, to utilize this potential, it is crucial to optimize their in vitro cultivation conditions. Three-dimensional (3D) culture methods using cell-laden hydrogels aim to mimic the physiological microenvironment in vitro, thus preserving MSC biological functionalities. Cellulosic hydrogels are particularly promising due to their biocompatibility, sustainability, and tunability in terms of chemical, morphological, and mechanical properties. This study investigated the impact of (1) two physical crosslinking scenarios for hydrogels derived from anionic cellulose nanofibers (to-CNF) used to encapsulate adipose-derived MSCs (adMSCs) and (2) physiological culture conditions on the in vitro proliferation, differentiation, and extracellular vesicle (EV) production of these adMSCs. The results revealed that additional Ca2+-mediated crosslinking, intended to complement the self-assembly and gelation of aqueous to-CNF in the adMSC cultivation medium, adversely affected both the mechanical properties of the hydrogel spheres and the growth of the encapsulated cells. However, cultivation under dynamic and hypoxic conditions significantly improved the proliferation and differentiation of the encapsulated adMSCs. Furthermore, it was demonstrated that the adMSCs in the CNF hydrogel spheres exhibited potential for scalable EV production with potent immunosuppressive capacities in a bioreactor system. These findings underscore the importance of physiological culture conditions and the suitability of cellulosic materials for enhancing the therapeutic potential of MSCs. Overall, this study provides valuable insights for optimizing the in vitro cultivation of MSCs for various applications, including tissue engineering, drug testing, and EV-based therapies.
Collapse
Affiliation(s)
- Ilias Nikolits
- Institute of Cell and Tissue Culture Technologies, Department of Biotechnology, University of Natural Resources and Life Sciences Vienna, Muthgasse 18, 1190 Vienna, Austria; (I.N.); (F.C.-P.)
| | - Farhad Chariyev-Prinz
- Institute of Cell and Tissue Culture Technologies, Department of Biotechnology, University of Natural Resources and Life Sciences Vienna, Muthgasse 18, 1190 Vienna, Austria; (I.N.); (F.C.-P.)
| | - Dominik Egger
- Institute of Cell Biology and Biophysics, Leibniz University Hannover, Herrenhäuser Strasse 2, 30419 Hannover, Germany;
| | - Falk Liebner
- Institute of Chemistry of Renewable Resources, Department of Chemistry, University of Natural Resources and Life Sciences Vienna, Konrad Lorenz Strasse 24, 3430 Tulln, Austria;
| | - Nicolas Mytzka
- MicroDiagnostics Unit, Fraunhofer Institute for Cell Therapy and Immunology, Perlickstraße 1, 04103 Leipzig, Germany;
| | - Cornelia Kasper
- Institute of Cell and Tissue Culture Technologies, Department of Biotechnology, University of Natural Resources and Life Sciences Vienna, Muthgasse 18, 1190 Vienna, Austria; (I.N.); (F.C.-P.)
| |
Collapse
|
32
|
Kong W, Bao Y, Li W, Guan D, Yin Y, Xiao Y, Zhu S, Sun Y, Xia Z. Collaborative Enhancement of Diabetic Wound Healing and Skin Regeneration by Recombinant Human Collagen Hydrogel and hADSCs. Adv Healthc Mater 2024:e2401012. [PMID: 39388509 DOI: 10.1002/adhm.202401012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 09/09/2024] [Indexed: 10/12/2024]
Abstract
Stem cell-based therapies hold significant promise for chronic wound healing and skin appendages regeneration, but challenges such as limited stem cell lifespan and poor biocompatibility of delivery systems hinder clinical application. In this study, an in situ delivery system for human adipose-derived stem cells is developed (hADSCs) to enhance diabetic wound healing. The system utilizes a photo-crosslinking recombinant human type III collagen (rHCIII) hydrogel to encapsulate hADSCs, termed the hADSCs@rHCIII hydrogel. This hydrogel undergoes local crosslinking at the wound site, establishing a sturdy 3D niche suitable for stem cell function. Consequently, the encapsulated hADSCs exhibit strong attachment and spreading within the hydrogels, maintaining their proliferation, metabolic activity, and viability for up to three weeks in vitro. Importantly, in vivo studies demonstrate that the hADSCs@rHCIII hydrogel achieves significant in situ delivery of stem cells, prolonging their retention within the wound. This ultimately enhances their immunomodulatory capabilities, promotes neovascularization and granulation tissue formation, facilitates matrix remodeling, and accelerates healing in a diabetic mouse wound model. Collectively, these findings highlight the potential of the conveniently-prepared and user-friendly hADSCs@rHCIII hydrogel as a promising therapeutic approach for diabetic wound treatment and in situ skin regeneration.
Collapse
Affiliation(s)
- Weishi Kong
- Department of Burn Surgery, the First Affiliated Hospital, Naval Medical University, Shanghai, 200433, P. R. China
- Research Unit of key techniques for treatment of burns and combined burns and trauma injury, Chinese Academy of Medical Sciences, Shanghai, 200433, P. R. China
| | - Yulu Bao
- Department of Burn Surgery, the First Affiliated Hospital, Naval Medical University, Shanghai, 200433, P. R. China
- Research Unit of key techniques for treatment of burns and combined burns and trauma injury, Chinese Academy of Medical Sciences, Shanghai, 200433, P. R. China
| | - Wei Li
- Department of Burn Surgery, the First Affiliated Hospital, Naval Medical University, Shanghai, 200433, P. R. China
- Research Unit of key techniques for treatment of burns and combined burns and trauma injury, Chinese Academy of Medical Sciences, Shanghai, 200433, P. R. China
| | - Dingding Guan
- Department of Burn Surgery, the First Affiliated Hospital, Naval Medical University, Shanghai, 200433, P. R. China
- Research Unit of key techniques for treatment of burns and combined burns and trauma injury, Chinese Academy of Medical Sciences, Shanghai, 200433, P. R. China
| | - Yating Yin
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, P. R. China
- Department of Burn and Plastic Surgery, Seventh People's Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200137, P. R. China
| | - Yongqiang Xiao
- Department of Burn Surgery, the First Affiliated Hospital, Naval Medical University, Shanghai, 200433, P. R. China
- Research Unit of key techniques for treatment of burns and combined burns and trauma injury, Chinese Academy of Medical Sciences, Shanghai, 200433, P. R. China
- ENT Institute, Department of Facial Plastic and Reconstructive Surgery, Eye & ENT Hospital, Fudan University, Shanghai, 200031, P. R. China
| | - Shihui Zhu
- Department of Burn Surgery, the First Affiliated Hospital, Naval Medical University, Shanghai, 200433, P. R. China
- Research Unit of key techniques for treatment of burns and combined burns and trauma injury, Chinese Academy of Medical Sciences, Shanghai, 200433, P. R. China
- Department of Burns and Plastic Surgery, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, P. R. China
| | - Yu Sun
- Department of Burn Surgery, the First Affiliated Hospital, Naval Medical University, Shanghai, 200433, P. R. China
- Research Unit of key techniques for treatment of burns and combined burns and trauma injury, Chinese Academy of Medical Sciences, Shanghai, 200433, P. R. China
| | - Zhaofan Xia
- Department of Burn Surgery, the First Affiliated Hospital, Naval Medical University, Shanghai, 200433, P. R. China
- Research Unit of key techniques for treatment of burns and combined burns and trauma injury, Chinese Academy of Medical Sciences, Shanghai, 200433, P. R. China
| |
Collapse
|
33
|
Singer J, Knezic N, Layne J, Gohring G, Christiansen J, Rothrauff B, Huard J. Enhancing Cartilage Repair: Surgical Approaches, Orthobiologics, and the Promise of Exosomes. Life (Basel) 2024; 14:1149. [PMID: 39337932 PMCID: PMC11432843 DOI: 10.3390/life14091149] [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: 07/28/2024] [Revised: 08/22/2024] [Accepted: 08/30/2024] [Indexed: 09/30/2024] Open
Abstract
Treating cartilage damage is challenging as its ability for self-regeneration is limited. Left untreated, it can progress to osteoarthritis (OA), a joint disorder characterized by the deterioration of articular cartilage and other joint tissues. Surgical options, such as microfracture and cell/tissue transplantation, have shown promise as techniques to harness the body's endogenous regenerative capabilities to promote cartilage repair. Nonetheless, these techniques have been scrutinized due to reported inconsistencies in long-term outcomes and the tendency for the defects to regenerate as fibrocartilage instead of the smooth hyaline cartilage native to joint surfaces. Orthobiologics are medical therapies that utilize biologically derived substances to augment musculoskeletal healing. These treatments are rising in popularity because of their potential to enhance surgical standards of care. More recent developments in orthobiologics have focused on the role of exosomes in articular cartilage repair. Exosomes are nano-sized extracellular vesicles containing cargo such as proteins, lipids, and nucleic acids, and are known to facilitate intercellular communication, though their regenerative potential still needs to be fully understood. This review aims to demonstrate the advancements in cartilage regeneration, highlight surgical and biological treatment options, and discuss the recent strides in understanding the precise mechanisms of action involved.
Collapse
Affiliation(s)
- Jacob Singer
- Linda and Mitch Hart Regenerative and Personalized Medicine, Steadman Philippon Research Institute, Vail, CO 81657, USA
| | - Noah Knezic
- Linda and Mitch Hart Regenerative and Personalized Medicine, Steadman Philippon Research Institute, Vail, CO 81657, USA
| | - Jonathan Layne
- Linda and Mitch Hart Regenerative and Personalized Medicine, Steadman Philippon Research Institute, Vail, CO 81657, USA
| | - Greta Gohring
- Linda and Mitch Hart Regenerative and Personalized Medicine, Steadman Philippon Research Institute, Vail, CO 81657, USA
| | - Jeff Christiansen
- Linda and Mitch Hart Regenerative and Personalized Medicine, Steadman Philippon Research Institute, Vail, CO 81657, USA
| | - Ben Rothrauff
- Linda and Mitch Hart Regenerative and Personalized Medicine, Steadman Philippon Research Institute, Vail, CO 81657, USA
| | - Johnny Huard
- Linda and Mitch Hart Regenerative and Personalized Medicine, Steadman Philippon Research Institute, Vail, CO 81657, USA
| |
Collapse
|
34
|
Su Y, Chen M, Xu W, Gu P, Fan X. Advances in Extracellular-Vesicles-Based Diagnostic and Therapeutic Approaches for Ocular Diseases. ACS NANO 2024; 18:22793-22828. [PMID: 39141830 PMCID: PMC11363148 DOI: 10.1021/acsnano.4c08486] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 07/30/2024] [Accepted: 08/02/2024] [Indexed: 08/16/2024]
Abstract
Extracellular vesicles (EVs) are nanoscale membrane vesicles of various sizes that can be secreted by most cells. EVs contain a diverse array of cargo, including RNAs, lipids, proteins, and other molecules with functions of intercellular communication, immune modulation, and regulation of physiological and pathological processes. The biofluids in the eye, including tears, aqueous humor, and vitreous humor, are important sources for EV-based diagnosis of ocular disease. Because the molecular cargos may reflect the biology of their parental cells, EVs in these biofluids, as well as in the blood, have been recognized as promising candidates as biomarkers for early diagnosis of ocular disease. Moreover, EVs have also been used as therapeutics and targeted drug delivery nanocarriers in many ocular disorders because of their low immunogenicity and superior biocompatibility in nature. In this review, we provide an overview of the recent advances in the field of EV-based studies on the diagnosis and therapeutics of ocular disease. We summarized the origins of EVs applied in ocular disease, assessed different methods for EV isolation from ocular biofluid samples, highlighted bioengineering strategies of EVs as drug delivery systems, introduced the latest applications in the diagnosis and treatment of ocular disease, and presented their potential in the current clinical trials. Finally, we briefly discussed the challenges of EV-based studies in ocular disease and some issues of concern for better focusing on clinical translational studies of EVs in the future.
Collapse
Affiliation(s)
- Yun Su
- Department
of Ophthalmology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
- Shanghai
Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai 200011, China
| | - Moxin Chen
- Department
of Ophthalmology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
- Shanghai
Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai 200011, China
| | - Wei Xu
- Department
of Ophthalmology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
- Shanghai
Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai 200011, China
| | - Ping Gu
- Department
of Ophthalmology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
- Shanghai
Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai 200011, China
| | - Xianqun Fan
- Department
of Ophthalmology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
- Shanghai
Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai 200011, China
| |
Collapse
|
35
|
Mainali BB, Yoo JJ, Ladd MR. Tissue engineering and regenerative medicine approaches in colorectal surgery. Ann Coloproctol 2024; 40:336-349. [PMID: 39228197 PMCID: PMC11375227 DOI: 10.3393/ac.2024.00437.0062] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 07/26/2024] [Accepted: 07/26/2024] [Indexed: 09/05/2024] Open
Abstract
Tissue engineering and regenerative medicine (TERM) is an emerging field that has provided new therapeutic opportunities by delivering innovative solutions. The development of nontraditional therapies for previously unsolvable diseases and conditions has brought hope and excitement to countless individuals globally. Many regenerative medicine therapies have been developed and delivered to patients clinically. The technology platforms developed in regenerative medicine have been expanded to various medical areas; however, their applications in colorectal surgery remain limited. Applying TERM technologies to engineer biological tissue and organ substitutes may address the current therapeutic challenges and overcome some complications in colorectal surgery, such as inflammatory bowel diseases, short bowel syndrome, and diseases of motility and neuromuscular function. This review provides a comprehensive overview of TERM applications in colorectal surgery, highlighting the current state of the art, including preclinical and clinical studies, current challenges, and future perspectives. This article synthesizes the latest findings, providing a valuable resource for clinicians and researchers aiming to integrate TERM into colorectal surgical practice.
Collapse
Affiliation(s)
- Bigyan B Mainali
- Department of General Surgery, Atrium Health Wake Forest Baptist, Winston-Salem, NC, USA
| | - James J Yoo
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC, USA
- Department of Biomedical Engineering, Wake Forest University, Winston-Salem, NC, USA
| | - Mitchell R Ladd
- Department of General Surgery, Atrium Health Wake Forest Baptist, Winston-Salem, NC, USA
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC, USA
- Department of Biomedical Engineering, Wake Forest University, Winston-Salem, NC, USA
| |
Collapse
|
36
|
Shi W, Zheng J, Zhang J, Dong X, Li Z, Xiao Y, Li Q, Huang X, Du Y. Desktop-Stereolithography 3D Printing of a Decellularized Extracellular Matrix/Mesenchymal Stem Cell Exosome Bioink for Vaginal Reconstruction. Tissue Eng Regen Med 2024; 21:943-957. [PMID: 38937423 PMCID: PMC11286906 DOI: 10.1007/s13770-024-00649-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 04/26/2024] [Accepted: 04/28/2024] [Indexed: 06/29/2024] Open
Abstract
BACKGROUND 3D-printing is widely used in regenerative medicine and is expected to achieve vaginal morphological restoration and true functional reconstruction. Mesenchymal stem cells-derived exosomes (MSCs-Exos) were applyed in the regeneration of various tissues. The current study aimed to explore the effctive of MSCs-Exos in vaginal reconstruction. METHODS In this work, hydrogel was designed using decellularized extracellular matrix (dECM) and gelatin methacrylate (GelMA) and silk fibroin (SF). The biological scaffolds were constructed using desktop-stereolithography. The physicochemical properties of the hydrogels were evaluated; Some experiments have been conducted to evaluate exosomes' effect of promotion vaginal reconstruction and to explore the mechanism in this process. RESULTS It was observed that the sustained release property of exosomes in the hydrogel both in vitro and in vitro.The results revealed that 3D scaffold encapsulating exosomes expressed significant effects on the vascularization and musule regeneration of the regenerative vagina tissue. Also, MSCs-Exos strongly promoted vascularization in the vaginal reconstruction of rats, which may through the PI3K/AKT signaling pathway. CONCLUSION The use of exosome-hydrogel composites improved the epithelial regeneration of vaginal tissue, increased angiogenesis, and promoted smooth muscle tissue regeneration. 3D-printed, lumenal scaffold encapsulating exosomes might be used as a cell-free alternative treatment strategy for vaginal reconstruction.
Collapse
Affiliation(s)
- Wenxin Shi
- Department of Obstetrics and Gynecology, The Second Hospital of Hebei Medical University, 215 Heping West Road, Shijiazhuang, 050000, Hebei, China
| | - Jiahua Zheng
- Department of Obstetrics and Gynecology, The Second Hospital of Hebei Medical University, 215 Heping West Road, Shijiazhuang, 050000, Hebei, China
| | - Jingkun Zhang
- Department of Obstetrics and Gynecology, The Second Hospital of Hebei Medical University, 215 Heping West Road, Shijiazhuang, 050000, Hebei, China
| | - Xiaoli Dong
- Department of Reproductive Medicine, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, Fujian, China
| | - Zhongkang Li
- Department of Obstetrics and Gynecology, The Second Hospital of Hebei Medical University, 215 Heping West Road, Shijiazhuang, 050000, Hebei, China
| | - Yanlai Xiao
- Department of Obstetrics and Gynecology, The Third Hospital of Hebei Medical University, 139 Ziqiang Road, Shijiazhuang, 050000, Hebei, China
| | - Qian Li
- Department of Obstetrics and Gynecology, The Second Hospital of Hebei Medical University, 215 Heping West Road, Shijiazhuang, 050000, Hebei, China
| | - Xianghua Huang
- Department of Obstetrics and Gynecology, The Second Hospital of Hebei Medical University, 215 Heping West Road, Shijiazhuang, 050000, Hebei, China.
| | - Yanfang Du
- Department of Obstetrics and Gynecology, The Second Hospital of Hebei Medical University, 215 Heping West Road, Shijiazhuang, 050000, Hebei, China.
| |
Collapse
|
37
|
Tang B, Xie X, Lu J, Huang W, Yang J, Tian J, Lei L. Designing biomaterials for the treatment of autoimmune diseases. APPLIED MATERIALS TODAY 2024; 39:102278. [DOI: 10.1016/j.apmt.2024.102278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2024]
|
38
|
Dai M, Lin X, Hua P, Wang S, Sun X, Tang C, Zhang C, Liu L. Antibacterial sequential growth factor delivery from alginate/gelatin methacryloyl microspheres for bone regeneration. Int J Biol Macromol 2024; 275:133557. [PMID: 38955293 DOI: 10.1016/j.ijbiomac.2024.133557] [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: 02/05/2024] [Revised: 06/13/2024] [Accepted: 06/28/2024] [Indexed: 07/04/2024]
Abstract
Autologous or allogeneic bone tissue grafts remain the mainstay of treatment for clinical bone defects. However, the risk of infection and donor scarcity in bone grafting pose challenges to the process. Therefore, the development of excellent biomaterial grafts is of great clinical importance for the repair of bone defects. In this study, we used gas-assisted microfluidics to construct double-cross-linked hydrogel microspheres with good biological function based on the ionic cross-linking of Cu2+ with alginate and photo-cross-linking of gelatin methacryloylamide (GelMA) by loading vascular endothelial growth factor (VEGF) and His-tagged bone morphogenetic protein-2 (BMP2) (AGMP@VEGF&BMP2). The Cu2+ component in the microspheres showed good antibacterial and drug-release behavior, whereas VEGF and BMP2 effectively promoted angiogenesis and bone tissue repair. In in vitro and in vivo experiments, the dual cross-linked hydrogel microspheres showed good biological function and biocompatibility. These results demonstrate that AGMP@VEGF&BMP2 microspheres could be used as a bone defect graft substitute to promote effective healing of bone defects and may be applied to other tissue engineering studies.
Collapse
Affiliation(s)
- Minghai Dai
- The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou 325200, China
| | - Xiufei Lin
- The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou 325200, China
| | - Peng Hua
- The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou 325200, China
| | - Simeng Wang
- The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou 325200, China
| | - Xiaoliang Sun
- The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou 325200, China
| | - Chengxuan Tang
- The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou 325200, China.
| | - Chi Zhang
- The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou 325200, China; Ruian Center of Chinese-American Research Institute for Diabetic Complications, Wenzhou 325200, China.
| | - Liangle Liu
- The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou 325200, China.
| |
Collapse
|
39
|
Jiang Y, Yusoff NM, Du J, Moses EJ, Lin JT. Current perspectives on mesenchymal stem cells as a potential therapeutic strategy for non-alcoholic fatty liver disease. World J Stem Cells 2024; 16:760-772. [PMID: 39086561 PMCID: PMC11287429 DOI: 10.4252/wjsc.v16.i7.760] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 05/18/2024] [Accepted: 06/14/2024] [Indexed: 07/25/2024] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) has emerged as a significant health challenge, characterized by its widespread prevalence, intricate natural progression and multifaceted pathogenesis. Although NAFLD initially presents as benign fat accumulation, it may progress to steatosis, non-alcoholic steatohepatitis, cirrhosis, and hepatocellular carcinoma. Mesenchymal stem cells (MSCs) are recognized for their intrinsic self-renewal, superior biocompatibility, and minimal immunogenicity, positioning them as a therapeutic innovation for liver diseases. Therefore, this review aims to elucidate the potential roles of MSCs in alleviating the progression of NAFLD by alteration of underlying molecular pathways, including glycolipid metabolism, inflammation, oxidative stress, endoplasmic reticulum stress, and fibrosis. The insights are expected to provide further understanding of the potential of MSCs in NAFLD therapeutics, and support the development of MSC-based therapy in the treatment of NAFLD.
Collapse
Affiliation(s)
- Yan Jiang
- School of Nursing, Xinxiang Medical University, Xinxiang 453000, Henan Province, China
- Department of Biomedical Sciences, Advanced Medical and Dental Institute, Universiti Sains Malaysia, Kepala Batas 13200, Pulau Pinang, Malaysia
| | - Narazah Mohd Yusoff
- Advanced Medical and Dental Institute, Universiti Sains Malaysia, Kepala Batas 13200, Pulau Pinang, Malaysia
| | - Jiang Du
- Henan Joint International Research Laboratory of Stem Cell Medicine, School of Medical Engineering, Xinxiang Medical University, Xinxiang 453003, Henan Province, China
- Stem Cells and Biotherapy Engineering Research Center of Henan, National Joint Engineering Laboratory of Stem Cells and Biotherapy, School of Life Science and Technology, Xinxiang Medical University, Xinxiang 453003, Henan Province, China
| | - Emmanuel Jairaj Moses
- Department of Biomedical Sciences, Advanced Medical and Dental Institute, Universiti Sains Malaysia, Kepala Batas 13200, Pulau Pinang, Malaysia
| | - Jun-Tang Lin
- Stem Cells and Biotherapy Engineering Research Center of Henan, National Joint Engineering Laboratory of Stem Cells and Biotherapy, School of Life Science and Technology, Xinxiang Medical University, Xinxiang 453003, Henan Province, China
- Henan Joint International Research Laboratory of Stem Cell Medicine, School of Medical Engineering, Xinxiang Medical University, Xinxiang 453000, Henan Province, China.
| |
Collapse
|
40
|
Liu X, Liu C, Lin Q, Shi T, Liu G. Exosome-loaded hydrogels for craniofacial bone tissue regeneration. Biomed Mater 2024; 19:052002. [PMID: 38815606 DOI: 10.1088/1748-605x/ad525c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Accepted: 05/30/2024] [Indexed: 06/01/2024]
Abstract
It is common for maladies and trauma to cause significant bone deterioration in the craniofacial bone, which can cause patients to experience complications with their appearance and their ability to function. Regarding grafting procedures' complications and disadvantages, the newly emerging field of tissue regeneration has shown promise. Tissue -engineered technologies and their applications in the craniofacial region are increasingly gaining prominence with limited postoperative risk and cost. MSCs-derived exosomes are widely applied in bone tissue engineering to provide cell-free therapies since they not only do not cause immunological rejection in the same way that cells do, but they can also perform a cell-like role. Additionally, the hydrogel system is a family of multipurpose platforms made of cross-linked polymers with considerable water content, outstanding biocompatibility, and tunable physiochemical properties for the efficient delivery of commodities. Therefore, the promising exosome-loaded hydrogels can be designed for craniofacial bone regeneration. This review lists the packaging techniques for exosomes and hydrogel and discusses the development of a biocompatible hydrogel system and its potential for exosome continuous delivery for craniofacial bone healing.
Collapse
Affiliation(s)
- Xiaojie Liu
- Department of Plastic Surgery, Yantaishan Hospital, Yantai, People's Republic of China
| | - Chang Liu
- Department of Plastic Surgery, Yantaishan Hospital, Yantai, People's Republic of China
| | - Qingquan Lin
- Institute of Applied Catalysis, College of Chemistry and Chemical Engineering, Yantai University, Yantai, People's Republic of China
| | - Ting Shi
- Department of Plastic Surgery, Yantaishan Hospital, Yantai, People's Republic of China
| | - Guanying Liu
- Department of Hand and Foot Surgery, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, People's Republic of China
| |
Collapse
|
41
|
Shahzad KA, Wang Z, Li X, Li J, Xu M, Tan F. Immunomodulatory effect of PLGA-encapsulated mesenchymal stem cells-derived exosomes for the treatment of allergic rhinitis. Front Immunol 2024; 15:1429442. [PMID: 39040099 PMCID: PMC11260627 DOI: 10.3389/fimmu.2024.1429442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 06/24/2024] [Indexed: 07/24/2024] Open
Abstract
Introduction Allergic rhinitis (AR) is an upper airway inflammatory disease of the nasal mucosa. Conventional treatments such as symptomatic pharmacotherapy and allergen-specific immunotherapy have considerable limitations and drawbacks. As an emerging therapy with regenerative potential and immunomodulatory effect, mesenchymal stem cell-derived exosomes (MSC-Exos) have recently been trialed for the treatment of various inflammatory and autoimmune diseases. Methods In order to achieve sustained and protected release of MSC-Exos for intranasal administration, we fabricated Poly(lactic-co-glycolic acid) (PLGA) micro and nanoparticles-encapsulated MSC-Exos (PLGA-Exos) using mechanical double emulsion for local treatment of AR. Preclinical in vivo imaging, ELISA, qPCR, flow cytometry, immunohistochemical staining, and multiomics sequencing were used for phenotypic and mechanistic evaluation of the therapeutic effect of PLGA-Exos in vitro and in vivo. Results The results showed that our PLGA platform could efficiently encapsulate and release the exosomes in a sustained manner. At protein level, PLGA-Exos treatment upregulated IL-2, IL-10 and IFN-γ, and downregulated IL-4, IL-17 and antigen-specific IgE in ovalbumin (OVA)-induced AR mice. At cellular level, exosomes treatment reduced Th2 cells, increased Tregs, and reestablished Th1/Th2 balance. At tissue level, PLGA-Exos significantly attenuated the infiltration of immune cells (e.g., eosinophils and goblet cells) in nasal mucosa. Finally, multiomics analysis discovered several signaling cascades, e.g., peroxisome proliferator-activated receptor (PPAR) pathway and glycolysis pathway, that might mechanistically support the immunomodulatory effect of PLGA-Exos. Discussion For the first time, we present a biomaterial-facilitated local delivery system for stem cell-derived exosomes as a novel and promising strategy for AR treatment.
Collapse
Affiliation(s)
- Khawar Ali Shahzad
- Department of ORL-HNS, Shanghai Fourth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
- Plasma Medicine and Surgical Implants Center, School of Medicine, Tongji University, Shanghai, China
| | - Zhao Wang
- Department of ORL-HNS, Shanghai Fourth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xuran Li
- Department of ORL-HNS, Shanghai Fourth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
- Plasma Medicine and Surgical Implants Center, School of Medicine, Tongji University, Shanghai, China
| | - Jiaojiao Li
- Department of ORL-HNS, Shanghai Fourth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
- Plasma Medicine and Surgical Implants Center, School of Medicine, Tongji University, Shanghai, China
| | - Maoxiang Xu
- Department of ORL-HNS, Shanghai Fourth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
- Plasma Medicine and Surgical Implants Center, School of Medicine, Tongji University, Shanghai, China
| | - Fei Tan
- Department of ORL-HNS, Shanghai Fourth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
- Plasma Medicine and Surgical Implants Center, School of Medicine, Tongji University, Shanghai, China
- The Royal College of Surgeons in Ireland, Dublin, Ireland
- The Royal College of Surgeons of England, London, United Kingdom
| |
Collapse
|
42
|
Rybachuk O, Nesterenko Y, Zhovannyk V. Modern advances in spinal cord regeneration: hydrogel combined with neural stem cells. Front Pharmacol 2024; 15:1419797. [PMID: 38994202 PMCID: PMC11236698 DOI: 10.3389/fphar.2024.1419797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 06/11/2024] [Indexed: 07/13/2024] Open
Abstract
Severe spinal cord injuries (SCI) lead to loss of functional activity of the body below the injury site, affect a person's ability to self-care and have a direct impact on performance. Due to the structural features and functional role of the spinal cord in the body, the consequences of SCI cannot be completely overcome at the expense of endogenous regenerative potential and, developing over time, lead to severe complications years after injury. Thus, the primary task of this type of injury treatment is to create artificial conditions for the regenerative growth of damaged nerve fibers through the area of the SCI. Solving this problem is possible using tissue neuroengineering involving the technology of replacing the natural tissue environment with synthetic matrices (for example, hydrogels) in combination with stem cells, in particular, neural/progenitor stem cells (NSPCs). This approach can provide maximum stimulation and support for the regenerative growth of axons of damaged neurons and their myelination. In this review, we consider the currently available options for improving the condition after SCI (use of NSC transplantation or/and replacement of the damaged area of the SCI with a matrix, specifically a hydrogel). We emphasise the expediency and effectiveness of the hydrogel matrix + NSCs complex system used for the reconstruction of spinal cord tissue after injury. Since such a complex approach (a combination of tissue engineering and cell therapy), in our opinion, allows not only to creation of conditions for supporting endogenous regeneration or mechanical reconstruction of the spinal cord, but also to strengthen endogenous regeneration, prevent the spread of the inflammatory process, and promote the restoration of lost reflex, motor and sensory functions of the injured area of spinal cord.
Collapse
Affiliation(s)
- Oksana Rybachuk
- Bogomoletz Institute of Physiology NAS of Ukraine, Kyiv, Ukraine
- Institute of Genetic and Regenerative Medicine, M. D. Strazhesko National Scientific Center of Cardiology, Clinical and Regenerative Medicine, National Academy of Medical Sciences of Ukraine, Kyiv, Ukraine
| | | | | |
Collapse
|
43
|
Mitić D, Čarkić J, Jaćimović J, Lazarević M, Jakšić Karišik M, Toljić B, Milašin J. The Impact of Nano-Hydroxyapatite Scaffold Enrichment on Bone Regeneration In Vivo-A Systematic Review. Biomimetics (Basel) 2024; 9:386. [PMID: 39056827 PMCID: PMC11274561 DOI: 10.3390/biomimetics9070386] [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: 05/14/2024] [Revised: 06/20/2024] [Accepted: 06/22/2024] [Indexed: 07/28/2024] Open
Abstract
OBJECTIVES In order to ensure improved and accelerated bone regeneration, nano-hydroxyapatite scaffolds are often enriched with different bioactive components to further accelerate and improve bone healing. In this review, we critically examined whether the enrichment of nHAp/polymer scaffolds with growth factors, hormones, polypeptides, microRNAs and exosomes improved new bone formation in vivo. MATERIALS AND METHODS Out of 2989 articles obtained from the literature search, 106 papers were read in full, and only 12 articles met the inclusion criteria for this review. RESULTS Several bioactive components were reported to stimulate accelerated bone regeneration in a variety of bone defect models, showing better results than bone grafting with nHAp scaffolds alone. CONCLUSIONS The results indicated that composite materials based on nHAp are excellent candidates as bone substitutes, while nHAp scaffold enrichment further accelerates bone regeneration. The standardization of animal models should be provided in order to clearly define the most significant parameters of in vivo studies. Only in this way can the adequate comparison of findings from different in vivo studies be possible, further advancing our knowledge on bone regeneration and enabling its translation to clinical settings.
Collapse
Affiliation(s)
- Dijana Mitić
- School of Dental Medicine, University of Belgrade, 11000 Belgrade, Serbia; (J.Č.); (J.J.); (M.L.); (M.J.K.); (B.T.); (J.M.)
| | | | | | | | | | | | | |
Collapse
|
44
|
Tang D, Tang W, Chen H, Liu D, Jiao F. Synergistic Effects of Icariin and Extracellular Vesicles Derived from Rabbit Synovial Membrane-Derived Mesenchymal Stem Cells on Osteochondral Repair via the Wnt/ β-Catenin Pathway. Anal Cell Pathol (Amst) 2024; 2024:1083143. [PMID: 38946863 PMCID: PMC11214593 DOI: 10.1155/2024/1083143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 05/24/2024] [Accepted: 06/03/2024] [Indexed: 07/02/2024] Open
Abstract
Objectives Osteochondral defects (OCDs) are localized areas of damaged cartilage and underlying subchondral bone that can produce pain and seriously impair joint function. Literature reports indicated that icariin (ICA) has the effect of promoting cartilage repair. However, its mechanism remains unclear. Here, we explored the effects of icariin and extracellular vesicles (EVs) from rabbit synovial-derived mesenchymal stem cells (rSMSCs) on repairing of OCDs. Materials and Methods Rabbit primary genicular chondrocytes (rPGCs), knee skeletal muscle cells (rSMCKs), and rSMSCs, and extracellular vesicles derived from the latter two cells (rSMCK-EVs and rSMSC-EVs) were isolated and identified. The rPGCs were stimulated with ICA, rSMSC-EVs either separately or in combination. The rSMCK-EVs were used as a control. After stimulation, chondrogenic-related markers were analyzed by quantitative RT-PCR and western blotting. Cell proliferation was determined by the CCK-8 assay. The preventative effects of ICA and SMSC-EVs in vivo were determined by H&E and toluidine blue staining. Immunohistochemical analyses were performed to evaluate the levels of COL2A1 and β-catenin in vivo. Results. In vitro, the proliferation of rPGCs was markedly increased by ICA treatment in a dose-dependent manner. When compared with ICA or rSMSC-EVs treatment alone, combined treatment with ICA and SMSC-EVs produced stronger stimulative effects on cell proliferation. Moreover, combined treatment with ICA and rSMSC-EVs promoted the expression of chondrogenic-related gene, including COL2A1, SOX-9, and RUNX2, which may be via the activation of the Wnt/β-catenin pathway. In vivo, combined treatment with rSMSC-EVs and ICA promoted cartilage repair in joint bone defects. Results also showed that ICA or rSMSC-EVs both promoted the COL2A1 and β-catenin protein accumulation in articular cartilage, and that was further enhanced by combined treatment with rSMSC-EVs and ICA. Conclusion Our findings highlight the promising potential of using combined treatment with ICA and rSMSC-EVs for promoting osteochondral repair.
Collapse
Affiliation(s)
- Dongming Tang
- Department of Joint SurgeryGuangzhou Hospital of Integrated Traditional and Western Medicine, Guangzhou, China
| | - Wang Tang
- Department of Spine SurgeryGuangzhou Hospital of Integrated Traditional and Western Medicine, Guangzhou, China
| | - Huanqing Chen
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Donghua Liu
- Department of Spine SurgeryGuangzhou Hospital of Integrated Traditional and Western Medicine, Guangzhou, China
| | - Feng Jiao
- Department of Joint SurgeryGuangzhou Hospital of Integrated Traditional and Western Medicine, Guangzhou, China
| |
Collapse
|
45
|
Wang Y, Shi X. The potential mechanisms and treatment effects of stem cell-derived exosomes in cardiac reengineering. NANOTECHNOLOGY 2024; 35:362005. [PMID: 38834043 DOI: 10.1088/1361-6528/ad53d1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 06/04/2024] [Indexed: 06/06/2024]
Abstract
Exosomes are extracellular vesicles of diverse compositions that are secreted by numerous cell types. Exosomes contain significant bioactive components, including lipids, proteins, mRNA, and miRNA. Exosomes play an important role in regulating cellular signaling and trafficking under both normal physiological and pathological circumstances. A multitude of factors, including thermal stress, ribosomal stress, endoplasmic reticulum stress, and oxidative stress influence the concentrations of exosomal mRNA, miRNA, proteins, and lipids. It has been stated that exosomes derived from stem cells (SCs) modulate a range of stresses by preventing or fostering cell balance. Exosomes derived from SCs facilitate recovery by facilitating cross-cellular communication via the transmission of information in the form of proteins, lipids, and other components. For this reason, exosomes are used as biomarkers to diagnose a wide variety of diseases. The focus of this review is the bioengineering of artificial exosomal cargoes. This process encompasses the control and transportation of particular exosomal cargoes, including but not limited to small molecules, recombinant proteins, immune modulators, and therapeutic medications. Therapeutic approaches of this nature have the potential to deliver therapeutic medications precisely to the intended site for the cure of a variety of disorders. Notably, our attention has been directed towards the therapeutic implementations of exosomes derived from SCs in the cure of cardiovascular ailments, including but not limited to ischemic heart disease, myocardial infarction, sepsis, heart failure, cardiomyopathy, and cardiac fibrosis. In general, researchers employ two methodologies when it comes to exosomal bioengineering. This review aims to explain the function of exosomes derived from SCs in the regulation of stress and present a novel therapeutic approach for cardiovascular disorders.
Collapse
Affiliation(s)
- Yibin Wang
- Department of Cardiology, Hangzhou Ninth People's Hospital, Hangzhou 311225, People's Republic of China
| | - Xiulian Shi
- Emergency Department, Chun'an First People's Hospital, Hangzhou 311700, People's Republic of China
| |
Collapse
|
46
|
Peng J, Yang T, Chen S, Deng N, Luo X, Liao R, Su B. Utilization of Hydrogels in Mesenchymal Stem Cell-Based Therapy for Kidney Diseases. TISSUE ENGINEERING. PART B, REVIEWS 2024; 30:315-326. [PMID: 37819717 DOI: 10.1089/ten.teb.2023.0196] [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: 10/13/2023]
Abstract
Kidney diseases are major global health problems, with high prevalence and mortality. However, current treatment strategies for kidney diseases fail to achieve satisfactory efficacy. Mesenchymal stem cell (MSC)-based therapy has been a promising strategy for treating kidney diseases. Preclinical studies have proven their safety and effectiveness in treating acute kidney injury (AKI) and chronic kidney disease (CKD), but the outcomes of clinical trials have shown very limited clinical efficacy. A variety of innovative approaches have been proposed to enhance the therapeutic potential of MSCs, and hydrogels are attractive candidates. Hydrogels are three-dimensional (3D) networks formed by hydrophilic polymers of natural or synthetic origin with diverse physical and chemical properties. They have been widely applied in the field of drug delivery and regenerative medicine, including MSC-based therapy. Many studies have proven that hydrogels can improve the therapeutic efficacy of MSCs for kidney diseases, but there are still challenges limiting the widespread application of this method. In this review, we introduce the application of MSCs in kidney diseases and the factors that influence therapeutic efficiency and focus on the beneficial effects of hydrogels in MSC-based therapy for AKI and CKD.
Collapse
Affiliation(s)
- Jing Peng
- Department of Nephrology, West China Hospital, Sichuan University, Chengdu, China
| | - Tinghang Yang
- Department of Nephrology, West China Hospital, Sichuan University, Chengdu, China
| | - Shanshan Chen
- Department of Nephrology, West China Hospital, Sichuan University, Chengdu, China
| | - Ningyue Deng
- Department of Nephrology, West China Hospital, Sichuan University, Chengdu, China
| | - Xinyao Luo
- Department of Nephrology, West China Hospital, Sichuan University, Chengdu, China
| | - Ruoxi Liao
- Department of Nephrology, West China Hospital, Sichuan University, Chengdu, China
| | - Baihai Su
- Department of Nephrology, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
47
|
He L, Zhang H, Zhao N, Liao L. A novel approach in biomedical engineering: The use of polyvinyl alcohol hydrogel encapsulating human umbilical cord mesenchymal stem cell-derived exosomes for enhanced osteogenic differentiation and angiogenesis in bone regeneration. Int J Biol Macromol 2024; 270:132116. [PMID: 38723803 DOI: 10.1016/j.ijbiomac.2024.132116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Revised: 04/09/2024] [Accepted: 05/04/2024] [Indexed: 05/19/2024]
Abstract
Developing effective methods for alveolar bone defect regeneration is a significant challenge in orthopedics. Exosomes from human umbilical cord mesenchymal stem cells (HUMSC-Exos) have shown potential in bone repair but face limitations due to undefined application methods and mechanisms. To address this, HUMSC-Exos were encapsulated in polyvinyl alcohol (PVA) hydrogel (Exo@PVA) to create a novel material for alveolar bone repair. This combination enhanced the osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs) and human umbilical vein endothelial cells (HUVECs) more effectively than Exos alone. Additionally, Exo@PVA significantly improved alveolar bone regeneration and defect repair in rats. The microRNA-21-5p (miR-21-5p) in Exo@PVA, identified through the GEO database and analyzed via in silico methods, played a crucial role. miR-21-5p promoted BMSC osteogenic differentiation by inhibiting WWP1-mediated KLF5 ubiquitination and enhanced HUVEC angiogenesis by targeting ATP2B4. These findings underscore the potential of an Exo-based approach with PVA hydrogel scaffolds for bone defect repair, operating through the miR-21-5p/WWP1/ATP2B4 signaling axis.
Collapse
Affiliation(s)
- Longlong He
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an 710004, PR China; Department of Implant Dentistry, College of Stomatology, Xi'an Jiaotong University, Xi'an 710004, PR China
| | - Hengwei Zhang
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an 710004, PR China
| | - Ningbo Zhao
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an 710004, PR China; Department of Implant Dentistry, College of Stomatology, Xi'an Jiaotong University, Xi'an 710004, PR China
| | - Lifan Liao
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an 710004, PR China; Department of Implant Dentistry, College of Stomatology, Xi'an Jiaotong University, Xi'an 710004, PR China.
| |
Collapse
|
48
|
Man J, Shen Y, Song Y, Yang K, Pei P, Hu L. Biomaterials-mediated radiation-induced diseases treatment and radiation protection. J Control Release 2024; 370:318-338. [PMID: 38692438 DOI: 10.1016/j.jconrel.2024.04.044] [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: 02/22/2024] [Revised: 03/31/2024] [Accepted: 04/25/2024] [Indexed: 05/03/2024]
Abstract
In recent years, the intersection of the academic and medical domains has increasingly spotlighted the utilization of biomaterials in radioactive disease treatment and radiation protection. Biomaterials, distinguished from conventional molecular pharmaceuticals, offer a suite of advantages in addressing radiological conditions. These include their superior biological activity, chemical stability, exceptional histocompatibility, and targeted delivery capabilities. This review comprehensively delineates the therapeutic mechanisms employed by various biomaterials in treating radiological afflictions impacting the skin, lungs, gastrointestinal tract, and hematopoietic systems. Significantly, these nanomaterials function not only as efficient drug delivery vehicles but also as protective agents against radiation, mitigating its detrimental effects on the human body. Notably, the strategic amalgamation of specific biomaterials with particular pharmacological agents can lead to a synergistic therapeutic outcome, opening new avenues in the treatment of radiation- induced diseases. However, despite their broad potential applications, the biosafety and clinical efficacy of these biomaterials still require in-depth research and investigation. Ultimately, this review aims to not only bridge the current knowledge gaps in the application of biomaterials for radiation-induced diseases but also to inspire future innovations and research directions in this rapidly evolving field.
Collapse
Affiliation(s)
- Jianping Man
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, Jiangsu 215123, China
| | - Yanhua Shen
- Experimental Animal Centre of Suzhou Medical College, Soochow University, Suzhou, Jiangsu 215005, China
| | - Yujie Song
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, Jiangsu 215123, China
| | - Kai Yang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, Jiangsu 215123, China
| | - Pei Pei
- Teaching and Research Section of Nuclear Medicine, School of Basic Medical Sciences, Anhui Medical University, 81 Meishan Road, Hefei 230032, Anhui, People's Republic of China..
| | - Lin Hu
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, Jiangsu 215123, China..
| |
Collapse
|
49
|
Mohammadi A, Shabani R, Bashiri Z, Rafiei S, Asgari H, Koruji M. Therapeutic potential of exosomes in spermatogenesis regulation and male infertility. Biol Cell 2024; 116:e2300127. [PMID: 38593304 DOI: 10.1111/boc.202300127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Revised: 02/19/2024] [Accepted: 02/22/2024] [Indexed: 04/11/2024]
Abstract
BACKGROUND Spermatogenesis is a fundamental process crucial for male reproductive health and fertility. Exosomes, small membranous vesicles released by various cell types, have recently garnered attention for their role in intercellular communication. OBJECTIVE This review aims to comprehensively explore the role of exosomes in regulating spermatogenesis, focusing on their involvement in testicular development and cell-to-cell communication. METHODS A systematic examination of literature was conducted to gather relevant studies elucidating the biogenesis, composition, and functions of exosomes in the context of spermatogenesis. RESULTS Exosomes play a pivotal role in orchestrating the complex signaling networks required for proper spermatogenesis. They facilitate the transfer of key regulatory molecules between different cell populations within the testes, including Sertoli cells, Leydig cells, and germ cells. CONCLUSION The emerging understanding of exosome-mediated communication sheds light on novel mechanisms underlying spermatogenesis regulation. Further research in this area holds promise for insights into male reproductive health and potential therapeutic interventions.
Collapse
Affiliation(s)
- Amirhossein Mohammadi
- Stem Cell and Regenerative Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Ronak Shabani
- Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Reproductive Sciences and Technology Research Center, Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Zahra Bashiri
- Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Endometrium and Endometriosis Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
- Omid Fertility & Infertility Clinic, Hamedan, Iran
| | - Sara Rafiei
- Department of Botany and Plant Sciences, Faculty of Biological Sciences, Alzahra University, Tehran, Iran
| | - Hamidreza Asgari
- Stem Cell and Regenerative Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Morteza Koruji
- Stem Cell and Regenerative Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
50
|
Bicer M. Revolutionizing dermatology: harnessing mesenchymal stem/stromal cells and exosomes in 3D platform for skin regeneration. Arch Dermatol Res 2024; 316:242. [PMID: 38795200 PMCID: PMC11127839 DOI: 10.1007/s00403-024-03055-4] [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: 11/28/2023] [Revised: 01/09/2024] [Accepted: 04/26/2024] [Indexed: 05/27/2024]
Abstract
Contemporary trends reveal an escalating interest in regenerative medicine-based interventions for addressing refractory skin defects. Conventional wound healing treatments, characterized by high costs and limited efficacy, necessitate a more efficient therapeutic paradigm to alleviate the economic and psychological burdens associated with chronic wounds. Mesenchymal stem/stromal cells (MSCs) constitute cell-based therapies, whereas cell-free approaches predominantly involve the utilization of MSC-derived extracellular vesicles or exosomes, both purportedly safe and effective. Exploiting the impact of MSCs by paracrine signaling, exosomes have emerged as a novel avenue capable of positively impacting wound healing and skin regeneration. MSC-exosomes confer several advantages, including the facilitation of angiogenesis, augmentation of cell proliferation, elevation of collagen production, and enhancement of tissue regenerative capacity. Despite these merits, challenges persist in clinical applications due to issues such as poor targeting and facile removal of MSC-derived exosomes from skin wounds. Addressing these concerns, a three-dimensional (3D) platform has been implemented to emend exosomes, allowing for elevated levels, and constructing more stable granules possessing distinct therapeutic capabilities. Incorporating biomaterials to encapsulate MSC-exosomes emerges as a favorable approach, concentrating doses, achieving intended therapeutic effectiveness, and ensuring continual release. While the therapeutic potential of MSC-exosomes in skin repair is broadly recognized, their application with 3D biomaterial scenarios remains underexplored. This review synthesizes the therapeutic purposes of MSCs and exosomes in 3D for the skin restoration, underscoring their promising role in diverse dermatological conditions. Further research may establish MSCs and their exosomes in 3D as a viable therapeutic option for various skin conditions.
Collapse
Affiliation(s)
- Mesude Bicer
- Department of Bioengineering, Faculty of Life and Natural Sciences, Abdullah Gul University, Kayseri, 38080, Turkey.
| |
Collapse
|