1
|
Ma N, Huang L, Zhou Q, Zhang X, Luo Q, Song G. Mechanical stretch promotes the migration of mesenchymal stem cells via Piezo1/F-actin/YAP axis. Exp Cell Res 2025; 446:114461. [PMID: 39988125 DOI: 10.1016/j.yexcr.2025.114461] [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: 09/22/2024] [Revised: 01/15/2025] [Accepted: 02/16/2025] [Indexed: 02/25/2025]
Abstract
Mesenchymal stem cells (MSCs) have self-renewal ability and the potential for multi-directional differentiation, and their clinical application has promising prospects, but improving the migration ability of MSCs in vivo is one of the challenges. We previously determined mechanical stretch at 1 Hz with 10 % strain for 8 h can significantly promote MSC migration, however, the molecular mechanism remains poorly understood. Here, we reported that the expression and activity of yes-associated protein (YAP) are upregulated after mechanical stretch. As a classical inhibitor of the YAP-TEAD activity and YAP protein, the treatment of verteporfin (VP) suppressed mechanical stretch-promoted MSC migration. We also observed F-actin polymerization after mechanical stretch. Next, we used Latrunculin A (Lat A), the most widely used reagent to depolymerize actin filaments, to treat MSCs and we found that Lat A treatment inhibits MSC migration by suppressing YAP expression and activity. In addition, the protein expression of Piezo1 was also upregulated after mechanical stretch. Knockdown of Piezo1 suppressed mechanical stretch-promoted MSC migration by restraining F-actin polymerization. Together, these findings demonstrate the role of Piezo1/F-actin/YAP signaling pathway in MSC migration under mechanical stretch, providing new experimental evidence for an in-depth understanding the mechanobiological mechanism of MSC migration.
Collapse
Affiliation(s)
- Ning Ma
- College of Bioengineering, Chongqing University, Chongqing, 400030, China
| | - Lei Huang
- College of Bioengineering, Chongqing University, Chongqing, 400030, China
| | - Qianxu Zhou
- College of Bioengineering, Chongqing University, Chongqing, 400030, China
| | - Xiaomei Zhang
- Department of Hematology and Oncology, Chongqing University Cancer Hospital, Chongqing, 400030, China
| | - Qing Luo
- College of Bioengineering, Chongqing University, Chongqing, 400030, China
| | - Guanbin Song
- College of Bioengineering, Chongqing University, Chongqing, 400030, China.
| |
Collapse
|
2
|
Huang S, Cheng C, Wang Z, Li R, Li W, Yu K, Shi Q, Xue M. Low-intensity-pulsed-ultrasound-treated menstrual-blood-derived mesenchymal stem cells repair endometrial injury by PI3K/AKT pathway inhibition. Reprod Biomed Online 2025; 50:104486. [PMID: 39979162 DOI: 10.1016/j.rbmo.2024.104486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 09/12/2024] [Accepted: 10/07/2024] [Indexed: 02/22/2025]
Abstract
RESEARCH QUESTION What is the effect and underlying mechanism of low-intensity-pulsed-ultrasound (LIPUS)-treated menstrual-blood-derived mesenchymal stem cells (MenSC) on intrauterine adhesions (IUA)? DESIGN First, MenSC were identified and exposed to LIPUS. The proliferation, migration, invasion, cytokine secretion, and ability to differentiate into human endometrial epithelial cells (HEEC) of LIPUS-treated MenSC were characterized. In vitro, human endometrial stromal cells (HESC) were treated with 10 ng/ml transforming growth factor-β1 (TGF-β1) to simulate IUA, and then co-cultured indirectly with LIPUS-treated MenSC. In vivo, IUA rat models were constructed and LIPUS-treated MenSC were transplanted into the uterus. The morphology, structure, and levels of fibrosis and repair-related factors of the uterus were detected. In the mechanism study, insulin-like growth factor-1 (IGF-1) was applied to verify whether the PI3K/AKT pathway participated in the repair of endometrial injury by LIPUS-treated MenSC. RESULTS In vitro, LIPUS treatment showed beneficial effects on MenSC by promoting cell proliferation and migration; inhibiting apoptosis; and enhancing the expression of epidermal growth factor, hepatocyte growth factor and vascular endothelial growth factor. It also facilitated the differentiation of MenSC into HEEC while reducing the level of fibrosis in TGF-β1-treated HESC by decreasing the concentrations of p-PI3K and p-AKT. However, these effects were reversed with the use of IGF-1. In vivo, transplantation of LIPUS-treated MenSC resulted in increased uterine length, width and weight. The transplanted cells also improved completeness of the endometrial structure, reduced inflammatory infiltration, increased endometrial thickness and gland abundance, and decreased endometrial fibrosis. Additionally, increased concentations of endometrial-repair-related proteins and receptivity-related markers were observed after transplantation of LIPUS-treated MenSC. CONCLUSION LIPUS-treated MenSC repaired endometrial injury by inhibiting the PI3K/AKT pathway.
Collapse
Affiliation(s)
- Shuangying Huang
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, China.; Department of Obstetrics, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Chunxia Cheng
- Department of Obstetrics, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Zhibiao Wang
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, China
| | - Ruizhen Li
- Department of Obstetrics, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Waixing Li
- Department of Obstetrics, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Kenan Yu
- Department of Obstetrics, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Qiuling Shi
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, China..
| | - Min Xue
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, China.; Department of Obstetrics, The Third Xiangya Hospital of Central South University, Changsha, China..
| |
Collapse
|
3
|
Jia W, Wang T, Chen F, Liu Z, Hou X, Cao W, Zhao X, Lu B, Hu Y, Dong Y, Zhou J, Zhou Z, Zhan W. Low-Intensity Pulsed Ultrasound Responsive Scaffold Promotes Intramembranous and Endochondral Ossification via Ultrasonic, Thermal, and Electrical Stimulation. ACS NANO 2025; 19:4422-4439. [PMID: 39901850 DOI: 10.1021/acsnano.4c13357] [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: 02/05/2025]
Abstract
Multiple physical stimuli are expected to produce a synergistic effect to promote bone tissue regeneration. Low-intensity pulsed ultrasound (LIPUS) has been clinically used in bone repair for the mechanical stimulation that it provides. In addition, LIPUS can also excite the biomaterials to generate other physical stimuli such as thermal or electrical stimuli. In this study, a scaffold based on decellularized adipose tissue (DAT) is established by incorporating polydopamine-modified multilayer black phosphorus nanosheets (pDA-mBP@DAT). Their effect on bone repair under LIPUS stimulation and the potential mechanisms are further investigated. This scaffold possesses piezoelectric properties and generates a mild thermogenic stimulus when stimulated by LIPUS. With superior properties, this scaffold is demonstrated to have good cytocompatibility in vitro and in vivo. Simultaneously, LIPUS promotes cell attachment, migration, and osteogenic differentiation in the pDA-mBP@DAT scaffold. Furthermore, the combined use of pDA-mBP@DAT and LIPUS significantly affects the regenerative effect in rat models of critical-sized calvarial defects. The possible mechanisms include promoting osteogenesis and neovascularization and activating the Piezo1. This study presents insight into speeding up bone regeneration by the synergistic combination of LIPUS and pDA-mBP@DAT scaffolds.
Collapse
Affiliation(s)
- Wanru Jia
- Department of Ultrasound, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, P.R. China
- College of Health Science and Technology, Shanghai Jiao Tong University School of Medicine, No. 227 Chongqing South Road, Shanghai 200025, P.R. China
| | - Tianlong Wang
- Department of Orthopedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Feng Chen
- Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Shanghai Stomatological Hospital & School of Stomatology, Fudan University, Shanghai 201102, P.R. China
| | - Zhiqing Liu
- Department of Orthopedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Xiaodong Hou
- Department of Orthopedics, First Affiliated Hospital of Kunming Medical University, Kunming Medical University, Kunming 650032, P.R. China
| | - Wentao Cao
- Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Shanghai Stomatological Hospital & School of Stomatology, Fudan University, Shanghai 201102, P.R. China
| | - Xinyu Zhao
- Department of Orthopedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Bingqiang Lu
- Department of Orthopedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Yan Hu
- Department of Ultrasound, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, P.R. China
| | - Yijie Dong
- Department of Ultrasound, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, P.R. China
- College of Health Science and Technology, Shanghai Jiao Tong University School of Medicine, No. 227 Chongqing South Road, Shanghai 200025, P.R. China
| | - Jianqiao Zhou
- Department of Ultrasound, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, P.R. China
- College of Health Science and Technology, Shanghai Jiao Tong University School of Medicine, No. 227 Chongqing South Road, Shanghai 200025, P.R. China
| | - Zifei Zhou
- Department of Orthopedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Weiwei Zhan
- Department of Ultrasound, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, P.R. China
- College of Health Science and Technology, Shanghai Jiao Tong University School of Medicine, No. 227 Chongqing South Road, Shanghai 200025, P.R. China
| |
Collapse
|
4
|
Wu L, Lu Y, Liu L, Wang J, Bai Y, Song J, Heng BC, Wu T, Ren Q, Li T, Xu M, Deng X, He Y, Liu Y, Zhang X. BaTiO 3 Doping Enhances Ultrasound-Driven Piezoelectric Bactericidal Effects of Fibrous Poly(L-Lactic Acid) Dressings to Accelerate Septic Wound Healing. ACS APPLIED MATERIALS & INTERFACES 2024; 16:67477-67490. [PMID: 39601666 DOI: 10.1021/acsami.4c17407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Bacterial invasion in infected skin wounds triggers inflammation and impedes healing. Current therapeutic strategies incorporating drug interventions within wound dressings often result in drug resistance and delayed healing. Here, we developed a comprehensive therapeutic modality integrating piezoelectric fibrous dressing with controlled ultrasound stimulation for efficient healing in an infected wound model. The electrospun fibrous dressings composed of barium titanate (BaTiO3) doped poly(L-lactic acid) (PLLA) possess improved piezoelectric properties due to the aligned structure and high crystallinity, which achieved superior bactericidal efficacy upon ultrasound-mechanical-electric conversion that results in the production of reactive oxygen species (ROS). There were 88.72% and 90.43% killing rates of Staphylococcus aureus and Escherichia coli respectively upon ultrasound stimulation without any need for exogenous drugs, and a wound closure rate of 95.5% within 10 days. The in vivo results confirmed that this dressing effectively shortened wound closure time by about 2 days, with a much-improved healing rate of 14% compared with previously reported therapeutic strategies. This was accompanied by reduced inflammation and increased re-epithelialization and angiogenesis. Hence, our synergistic treatment by piezoelectric materials and controlled ultrasound stimulation provides a drug-free alternative approach in regenerative tissue engineering for simultaneously enhancing antibacterial effects and promoting wound healing.
Collapse
Affiliation(s)
- Liping Wu
- Institute of Medical Technology, Peking University Health Science Center, Beijing 100191, PR China
- Department of Dental Materials & Dental Medical Devices Testing Center, Peking University School and Hospital of Stomatology, Beijing 100081, PR China
| | - Yanhui Lu
- Department of Dental Materials & Dental Medical Devices Testing Center, Peking University School and Hospital of Stomatology, Beijing 100081, PR China
| | - Lulu Liu
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing 100081, PR China
| | - Jianfeng Wang
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun 130012, PR China
| | - Yunyang Bai
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing 100081, PR China
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, NMPA Key Laboratory for Dental Materials, Beijing Laboratory of Biomedical Materials & Beijing Key Laboratory of Digital Stomatology, Peking University School and Hospital of Stomatology, Beijing 100081, PR China
| | - Jia Song
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing 100081, PR China
| | - Boon Chin Heng
- Department of Dental Materials & Dental Medical Devices Testing Center, Peking University School and Hospital of Stomatology, Beijing 100081, PR China
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, NMPA Key Laboratory for Dental Materials, Beijing Laboratory of Biomedical Materials & Beijing Key Laboratory of Digital Stomatology, Peking University School and Hospital of Stomatology, Beijing 100081, PR China
| | - Tingting Wu
- Oral Translational Medicine Research Center, Shanxi Key Laboratory of Oral and Maxillofacial Repair, Reconstruction and Regeneration Joint Training Base, The First People's Hospital of Jinzhong, Jinzhong 030600, Shanxi Province, PR China
| | - Qiaomei Ren
- Oral Translational Medicine Research Center, Shanxi Key Laboratory of Oral and Maxillofacial Repair, Reconstruction and Regeneration Joint Training Base, The First People's Hospital of Jinzhong, Jinzhong 030600, Shanxi Province, PR China
| | - Tingjun Li
- Oral Translational Medicine Research Center, Shanxi Key Laboratory of Oral and Maxillofacial Repair, Reconstruction and Regeneration Joint Training Base, The First People's Hospital of Jinzhong, Jinzhong 030600, Shanxi Province, PR China
| | - Mingming Xu
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing 100081, PR China
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, NMPA Key Laboratory for Dental Materials, Beijing Laboratory of Biomedical Materials & Beijing Key Laboratory of Digital Stomatology, Peking University School and Hospital of Stomatology, Beijing 100081, PR China
| | - Xuliang Deng
- Institute of Medical Technology, Peking University Health Science Center, Beijing 100191, PR China
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing 100081, PR China
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, NMPA Key Laboratory for Dental Materials, Beijing Laboratory of Biomedical Materials & Beijing Key Laboratory of Digital Stomatology, Peking University School and Hospital of Stomatology, Beijing 100081, PR China
| | - Ying He
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing 100081, PR China
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, NMPA Key Laboratory for Dental Materials, Beijing Laboratory of Biomedical Materials & Beijing Key Laboratory of Digital Stomatology, Peking University School and Hospital of Stomatology, Beijing 100081, PR China
| | - Yang Liu
- Department of Dental Materials & Dental Medical Devices Testing Center, Peking University School and Hospital of Stomatology, Beijing 100081, PR China
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, NMPA Key Laboratory for Dental Materials, Beijing Laboratory of Biomedical Materials & Beijing Key Laboratory of Digital Stomatology, Peking University School and Hospital of Stomatology, Beijing 100081, PR China
- Oral Translational Medicine Research Center, Shanxi Key Laboratory of Oral and Maxillofacial Repair, Reconstruction and Regeneration Joint Training Base, The First People's Hospital of Jinzhong, Jinzhong 030600, Shanxi Province, PR China
| | - Xuehui Zhang
- Institute of Medical Technology, Peking University Health Science Center, Beijing 100191, PR China
- Department of Dental Materials & Dental Medical Devices Testing Center, Peking University School and Hospital of Stomatology, Beijing 100081, PR China
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, NMPA Key Laboratory for Dental Materials, Beijing Laboratory of Biomedical Materials & Beijing Key Laboratory of Digital Stomatology, Peking University School and Hospital of Stomatology, Beijing 100081, PR China
- Oral Translational Medicine Research Center, Shanxi Key Laboratory of Oral and Maxillofacial Repair, Reconstruction and Regeneration Joint Training Base, The First People's Hospital of Jinzhong, Jinzhong 030600, Shanxi Province, PR China
| |
Collapse
|
5
|
Li X, Deng Z, Lu W. Mechanistic study of the effect of flexible fixation and load-bearing stress environment on fracture healing and shaping. Animal Model Exp Med 2024; 7:816-823. [PMID: 38978345 PMCID: PMC11680484 DOI: 10.1002/ame2.12448] [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: 12/18/2023] [Revised: 04/27/2024] [Accepted: 05/22/2024] [Indexed: 07/10/2024] Open
Abstract
BACKGROUND The biomechanical environment created by suture-button fixation Latarjet is conducive to the healing and shaping of the transplanted coracoid, but its mechanism remains unclear. The latest research has found that the absence of stem cell chemokine (CXCL12) impeded bone regeneration in Sonic Hedgehog (SHH)-deficient animals. However, whether the biomechanical environment affects SHH and CXCL12 function has not been studied. METHODS Rat fracture models were constructed to simulate stress environments under non-load-bearing and load-bearing conditions. The fracture healing and shaping, as well as the expression levels of SHH and CXCL12, were assessed through gross viewing, micro-computed tomography (micro-CT), and histochemical staining. RESULTS Under flexible fixation, the relative bone volume (BV/TV) of rats exposed to the load-bearing stress environment was significantly higher than that of rats under a non-load-bearing stress environment (p ≤ 0.05). Adverse bone shaping was not observed in rats subjected to flexible fixation. The levels of SHH and CXCL12 in load-bearing rats exhibited significant elevation (p ≤ 0.05). Under a load-bearing stress environment, no significant difference was observed in the BV/TV between the flexible fixation group and the rigid fixation group (p ≥ 0.05), but there was excessive hyperplasia of the fracture callus in the rigid fixation group. The levels of SHH and CXCL12 in rats subjected to rigid fixation were significantly elevated (p ≤ 0.05). CONCLUSIONS Flexible fixation and load-bearing stress environment may contribute to bone healing and shaping by influencing the levels of SHH and CXCL12, suggested that this mechanism may be relevant to the healing and shaping of the transplanted coracoid after suture-button fixation Latarjet.
Collapse
Affiliation(s)
- Xingfu Li
- Department of Sports MedicineShenzhen Second People's Hospital (The First Affiliated Hospital of Shenzhen University)ShenzhenGuangdongChina
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National‐Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical EngineeringShenzhen University Medical SchoolShenzhenGuangdongChina
| | - Zhenhan Deng
- Department of Sports MedicineShenzhen Second People's Hospital (The First Affiliated Hospital of Shenzhen University)ShenzhenGuangdongChina
- Department of Orthopedics SurgeryThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouZhejiangChina
| | - Wei Lu
- Department of Sports MedicineShenzhen Second People's Hospital (The First Affiliated Hospital of Shenzhen University)ShenzhenGuangdongChina
| |
Collapse
|
6
|
Xie B, Ma H, Yang F, Chen H, Guo Y, Zhang H, Li T, Huang X, Zhao Y, Li X, Du J. Development and evaluation of 3D composite scaffolds with piezoelectricity and biofactor synergy for enhanced articular cartilage regeneration. J Mater Chem B 2024; 12:10416-10433. [PMID: 39291892 DOI: 10.1039/d4tb01319k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
The inability of articular cartilage to self-repair following injuries frequently precipitates osteoarthritis, profoundly affecting patients' quality of life. Given the limitations inherent in current clinical interventions, an urgent need exists for more effective cartilage regeneration methodologies. Previous studies have underscored the potential of electrical stimulation in cartilage repair, thus motivating the investigation of innovative strategies. The present study introduces a three-dimensional scaffold fabricated through a composite technique that leverages the synergy between piezoelectricity and biofactors to enhance cartilage repair. This scaffold is composed of polylactic acid (PLLA) and barium titanate (BT) for piezoelectric stimulation and at the bottom with a collagen-coated layer infused with fibroblast growth factor-18 (FGF-18) for biofactor delivery. Designed to emulate the properties of natural cartilage, the scaffold enables controlled generation of piezoelectric charges and the sustained release of biofactors. In vitro tests confirm that the scaffold promotes chondrocyte proliferation, matrix hyperplasia, cellular migration, and the expression of genes associated with cartilage formation. Moreover, in vivo studies on rabbits have illustrated its efficacy in catalyzing the in situ regeneration of articular cartilage defects and remodeling the extracellular matrix. This innovative approach offers significant potential for enhancing cartilage repair and holds profound implications for regenerative medicine.
Collapse
Affiliation(s)
- Bowen Xie
- Department of Orthopedics, Air Force Medical Center, Beijing 100142, China.
- Air Force Clinical College, The Fifth School of Clinical Medicine, Anhui Medical University, Hefei 230032, China.
| | - Hebin Ma
- Medical School of the PLA General Hospital, Beijing 100853, China
- Senior Department of Orthopedics, The Fourth Medical Center of the PLA General Hospital, Beijing 100048, China.
| | - Fengyuan Yang
- Graduate School of Medicine, China Medical University, Shenyang 110122, China
| | - Hongguang Chen
- Senior Department of Orthopedics, The Fourth Medical Center of the PLA General Hospital, Beijing 100048, China.
| | - Ya'nan Guo
- Senior Department of Orthopedics, The Fourth Medical Center of the PLA General Hospital, Beijing 100048, China.
| | - Hongxing Zhang
- Department of Orthopedics, Air Force Medical Center, Beijing 100142, China.
| | - Tengfei Li
- Department of Orthopedics, Air Force Medical Center, Beijing 100142, China.
| | - Xiaogang Huang
- Department of Orthopedics, Air Force Medical Center, Beijing 100142, China.
| | - Yantao Zhao
- Senior Department of Orthopedics, The Fourth Medical Center of the PLA General Hospital, Beijing 100048, China.
| | - Xiaojie Li
- Department of Orthopedics, Air Force Medical Center, Beijing 100142, China.
| | - Junjie Du
- Department of Orthopedics, Air Force Medical Center, Beijing 100142, China.
- Air Force Clinical College, The Fifth School of Clinical Medicine, Anhui Medical University, Hefei 230032, China.
- Graduate School of Medicine, China Medical University, Shenyang 110122, China
| |
Collapse
|
7
|
Xiong H, Cao M, Yu Y, Duan X, Sun L, Tang L, Fan X. Study on the Effects of Low-Intensity Pulsed Ultrasound and Iron Ions for Proliferation and Differentiation of Osteoblasts. ULTRASOUND IN MEDICINE & BIOLOGY 2024:S0301-5629(24)00265-5. [PMID: 39209558 DOI: 10.1016/j.ultrasmedbio.2024.07.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Revised: 06/25/2024] [Accepted: 07/08/2024] [Indexed: 09/04/2024]
Abstract
OBJECTIVE This study involved the proliferation and differentiation of osteoblasts treated with low-intensity pulsed ultrasound (LIPUS) and iron (Fe3+) ions, respectively. The biological effects of LIPUS and Fe3+ ions on the proliferation and differentiation of osteoblasts were also evaluated. METHODS MC3T3-E1 cells were seeded in six-well plates with the medium, which contained different concentrations of Fe3+ (0, 100, 200, 300, 400, 500, 600 and 700 μg L-1, respectively). LIPUS treatment was directed at the bottom of the plate for 20 min at an intensity of 80 mW cm-2 every day. RESULTS Viability results showed that a dose of 400 μg L-1 Fe3+ ions had the best effect at promoting osteogenic proliferation in cell culture. The results of alkaline phosphatase staining and mineralization indicated that the differentiation of osteoblasts was promoted by LIPUS and Fe3+ ions. Fluorescence staining results showed that the number of cell nuclei in the LIPUS, Fe3+ and LIPUS-Fe groups increased by 37.20%, 55.81% and 89.76%, respectively. Migration data indicated that migration and proliferation rates were increased by LIPUS and Fe3+, and the results of protein expression indicated that LIPUS and Fe3+ may increase the expression of Wnt, β-catenin, and Runx2, hence promoting normal bone regeneration and development. CONCLUSION The combination of LIPUS (1.5 MHz, 80 mW cm-2) and Fe3+ accelerates the proliferation and differentiation of osteoblasts significantly compared with single-factor treatment (stimulated by LIPUS and Fe3+ ions, respectively). This study could establish a foundation for LIPUS-responsive biomaterials in the repair and regeneration of bone tissues.
Collapse
Affiliation(s)
- Huanbin Xiong
- Institute of Sports Biology, Shaanxi Normal University, Xi'an, China
| | - Mengshu Cao
- Institute of Sports Biology, Shaanxi Normal University, Xi'an, China
| | - Yanan Yu
- Institute of Sports Biology, Shaanxi Normal University, Xi'an, China
| | - Xueyou Duan
- Institute of Sports Biology, Shaanxi Normal University, Xi'an, China
| | - Lijun Sun
- Institute of Sports Biology, Shaanxi Normal University, Xi'an, China
| | - Liang Tang
- Institute of Sports Biology, Shaanxi Normal University, Xi'an, China
| | - Xiushan Fan
- Institute of Sports Biology, Shaanxi Normal University, Xi'an, China.
| |
Collapse
|
8
|
Kim D, Lee MJ, Arai Y, Ahn J, Lee GW, Lee SH. Ultrasound-triggered three dimensional hyaluronic acid hydrogel promotes in vitro and in vivo reprogramming into induced pluripotent stem cells. Bioact Mater 2024; 38:331-345. [PMID: 38764447 PMCID: PMC11101682 DOI: 10.1016/j.bioactmat.2024.05.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 04/12/2024] [Accepted: 05/05/2024] [Indexed: 05/21/2024] Open
Abstract
Cellular reprogramming technologies have been developed with different physicochemical factors to improve the reprogramming efficiencies of induced pluripotent stem cells (iPSCs). Ultrasound is a clinically applied noncontact biophysical factor known for regulating various cellular behaviors but remains uninvestigated for cellular reprogramming. Here, we present a new reprogramming strategy using low-intensity ultrasound (LIUS) to improve cellular reprogramming of iPSCs in vitro and in vivo. Under 3D microenvironment conditions, increased LIUS stimulation shows enhanced cellular reprogramming of the iPSCs. The cellular reprogramming process facilitated by LIUS is accompanied by increased mesenchymal to epithelial transition and histone modification. LIUS stimulation transiently modulates the cytoskeletal rearrangement, along with increased membrane fluidity and mobility to increase HA/CD44 interactions. Furthermore, LIUS stimulation with HA hydrogel can be utilized in application of both human cells and in vivo environment, for enhanced reprogrammed cells into iPSCs. Thus, LIUS stimulation with a combinatorial 3D microenvironment system can improve cellular reprogramming in vitro and in vivo environments, which can be applied in various biomedical fields.
Collapse
Affiliation(s)
| | | | - Yoshie Arai
- Department of Biomedical Engineering, Dongguk University-Seoul, 04620, Seoul, South Korea
| | - Jinsung Ahn
- Department of Biomedical Engineering, Dongguk University-Seoul, 04620, Seoul, South Korea
| | - Gun Woo Lee
- Department of Biomedical Engineering, Dongguk University-Seoul, 04620, Seoul, South Korea
| | - Soo-Hong Lee
- Department of Biomedical Engineering, Dongguk University-Seoul, 04620, Seoul, South Korea
| |
Collapse
|
9
|
Altememy D, Kashani MHG, Fateme A, Khosravian P. New method to induce neurotrophin gene expression in human adipose-derived stem cells in vitro. J Adv Pharm Technol Res 2024; 15:214-219. [PMID: 39290551 PMCID: PMC11404434 DOI: 10.4103/japtr.japtr_390_23] [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: 08/08/2023] [Revised: 01/10/2024] [Accepted: 02/05/2024] [Indexed: 09/19/2024] Open
Abstract
Rosemary leaf extract, a well-known medicinal plant, can induce neurotrophin gene expression and proliferation in stem cells. Human adipose-derived stem cells (hASCs) with high proliferation and differentiation capacity are easily accessible and can be extracted with the least damage. This study evaluated the effect of rosemary extract (RE) on neurotrophin gene expression at 48 h postinduction in hASCs. hASCs were isolated from healthy female donors, aged 28-35 years, who had undergone abdominal liposuction. Passage-4 stem cells were cultured and treated with different doses of RE (from 30 to 70 µg/ml) containing 40% carnosic acid for 48 h. Reverse transcription-polymerase chain reaction was used to check the expression of neurotrophin genes. The expression of NTF3, NTF4, and nerve growth factor genes in cells treated with 40-60 µg/ml and the expression of GDNF in cells treated with 50-70 µg/ml of RE for 48 h showed a significant increase compared to cells cultured in serum-containing medium. However, different doses of RE showed no effect on brain-derived neurotrophic factor gene expression in the treated cells. RE (50, 60 µg/ml) leads to an increase of neurotrophin gene expression in hASCs as compared to routine cell culture. Hence, this protocol can be used to prepare ideal cell sources for cell therapy.
Collapse
Affiliation(s)
- Dhiya Altememy
- Department of Pharmaceutics, College of Pharmacy, Al-Zahraa University for Women, Karbala, Iraq
| | - Maryam Haji Ghasem Kashani
- Department of Cellular and Molecular Biology, School of Biology, Institute of Biological Sciences, Damghan University, Damghan, Iran
| | - Amirahmadi Fateme
- Department of Cellular and Molecular Biology, School of Biology, Institute of Biological Sciences, Damghan University, Damghan, Iran
| | - Pegah Khosravian
- Medical Plants Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| |
Collapse
|
10
|
Chen B, Zhu Q, Duan M, Li Q, Wang G, Guan X, Yu P, Xu X, He Y, Xu Y. Optimal Treatment Parameters for Ultrasound-Stimulated Microbubbles in Upregulating Proliferation and Stemness of Bone Marrow Mesenchymal Stem Cells. JOURNAL OF ULTRASOUND IN MEDICINE : OFFICIAL JOURNAL OF THE AMERICAN INSTITUTE OF ULTRASOUND IN MEDICINE 2024; 43:1333-1342. [PMID: 38563453 DOI: 10.1002/jum.16457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Revised: 03/16/2024] [Accepted: 03/23/2024] [Indexed: 04/04/2024]
Abstract
OBJECTIVES Ultrasound-targeted microbubble disruption (UTMD) is a widely used technique to improve the differentiation and proliferation capacity of mesenchymal stem cells (MSCs), but the optimal therapeutic parameters for UTMD are unclear. In this study, we aimed to find the appropriate peak negative pressure (PNP), which is a key parameter for enhancing the stemness properties and proliferation of MSCs. METHODS Experiments were performed in UTMD group, ultrasound (US) group under different PNP exposure conditions (0.5, 1.0, and 1.5 MPa), and control group. Apoptosis safety was analyzed by flow cytometry and MSC proliferation was measured at 12, 24, and 36 hours after irradiation by cell counting kit 8. The expression of the stemness genes NANOG, OCT-4, and SOX-2 were determined by enzyme-linked immunosorbent assay (ELISA) or reverse transcription polymerase chain reaction. RESULTS The results showed that the 1.5 MPa UTMD-treated group had the highest proliferation capacity of MSCs at 24 hours. ELISA or quantitative reverse transcription polymerase chain reaction results showed that UTMD treatment of the 1.5 MPa group significantly upregulated the expression of the stemness genes NANOG, SOX-2, and OCT-4. CONCLUSIONS In conclusion, the appropriate peak PNP value of UTMD was 1.5 MPa, and 1.5 MPa-mediated UTMD group obviously promoted MSCs proliferation and maintained stemness by upregulating the expression of stemness genes.
Collapse
Affiliation(s)
- Beibei Chen
- Department of Ultrasound, Xinqiao Hospital, Army Medical University, Chongqing, China
- Department of Ultrasound, Postgraduate Training Basement of Jinzhou Medical University, The PLA Rocket Force Characteristic Medical Center, Beijing, China
| | - Qiong Zhu
- Department of Ultrasound, Xinqiao Hospital, Army Medical University, Chongqing, China
- Department of Ultrasound, 953th Hospital, Shigatse Branch, Xinqiao Hospital, Army Medical University, Shigatse, China
| | - Mao Duan
- Department of Ultrasound, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Qinglong Li
- Department of Ultrasound, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Gong Wang
- Department of Ultrasound, Postgraduate Training Basement of Jinzhou Medical University, The PLA Rocket Force Characteristic Medical Center, Beijing, China
| | - Xue Guan
- Department of Ultrasound, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Pu Yu
- Department of Ultrasound, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Xiaoxun Xu
- Department of Ultrasound, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Ying He
- Department of Ultrasound, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Yali Xu
- Department of Ultrasound, Xinqiao Hospital, Army Medical University, Chongqing, China
| |
Collapse
|
11
|
Hu X, Duan H, Zou D, Dong C, Wang Y, Wang Y, Li Z, Li Z. Acoustic vibration promotes in vitro expansion of human embryonic stem cells. AMERICAN JOURNAL OF STEM CELLS 2024; 13:143-151. [PMID: 39021373 PMCID: PMC11249672 DOI: 10.62347/pjfc2708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 05/06/2024] [Indexed: 07/20/2024]
Abstract
OBJECTIVES This study aimed to investigate the effect of acoustic vibration on the pluripotency of human embryonic stem cells (hESCs) and evaluate cell proliferation and self-renewal ability post-treatment. METHODS The human ES cell line H1 was used for the experiments. hESCs were treated with an acoustic vibration device. Their proliferative ability was subsequently detected using a colony formation assay, while the expression of pluripotency-related markers was detected via immunofluorescence staining. Finally, changes in gene expression levels were examined using quantitative polymerase chain reaction (qPCR) in the presence of appropriate primers. RESULTS Compared with normal cells in the control group, the morphology of experimental cells subjected to acoustic vibration did not significantly change. Contrastingly, the colony-forming efficiency of the experimental cells significantly increased. Immunofluorescence staining results showed the cells in experimental group were positive for the pluripotency markers NANOG, octamer-binding transcription factor 4 gene (OCT4), and SRY (sex determining region Y)-box 2 (SOX2). In addition, the expression levels of pluripotency genes NANOG, OCT4, SOX2, and Yes-associated protein (YAP)-related genes were up-regulated following acoustic vibration. CONCLUSIONS Our results revealed that acoustic vibration enhanced the proliferative ability of hESCs and increased the expression levels of NANOG, OCT4, SOX2, and YAP-related genes, indicating that acoustic vibration can optimize the self-renewal ability of hESCs and that the YAP signaling pathway may play a critical role in the functional process of acoustic vibration.
Collapse
Affiliation(s)
- Xiangyue Hu
- Shandong First Medical University (Shandong Academy of Medical Sciences)Jinan 250000, Shandong, China
- Eye Institute of Shandong First Medical University, State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of OphthalmologyQingdao 266071, Shandong, China
| | - Haoyun Duan
- Eye Institute of Shandong First Medical University, State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of OphthalmologyQingdao 266071, Shandong, China
| | - Dulei Zou
- Eye Institute of Shandong First Medical University, State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of OphthalmologyQingdao 266071, Shandong, China
- Eye Institute of Shandong First Medical University, Eye Hospital of Shandong First Medical University (Shandong Eye Hospital)Jinan 250021, Shandong, China
| | - Chunxiao Dong
- Eye Institute of Shandong First Medical University, State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of OphthalmologyQingdao 266071, Shandong, China
- Eye Institute of Shandong First Medical University, Eye Hospital of Shandong First Medical University (Shandong Eye Hospital)Jinan 250021, Shandong, China
| | - Yani Wang
- Eye Institute of Shandong First Medical University, State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of OphthalmologyQingdao 266071, Shandong, China
| | - Yao Wang
- Eye Institute of Shandong First Medical University, State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of OphthalmologyQingdao 266071, Shandong, China
| | - Zongren Li
- 970 Hospital of Chinese PLA Joint Logistic Support ForceWeihai 264200, Shandong, China
| | - Zongyi Li
- Shandong First Medical University (Shandong Academy of Medical Sciences)Jinan 250000, Shandong, China
- Eye Institute of Shandong First Medical University, State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of OphthalmologyQingdao 266071, Shandong, China
| |
Collapse
|
12
|
Wu J, Wei YT. [Regenerative rehabilitation: the effects of physical factors on regeneration]. ZHONGHUA SHAO SHANG YU CHUANG MIAN XIU FU ZA ZHI 2024; 40:301-306. [PMID: 38664023 DOI: 10.3760/cma.j.cn501225-20231229-00278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 05/01/2024]
Abstract
Wound regeneration and repair is one of the primary research fields in burn and wound repair surgery. In recent years, with the continuous advancement of treatment concept and technologies in the field of rehabilitation, the connection between rehabilitation treatment and wound regeneration and repair has become closer, forming a new concept "regenerative rehabilitation". This article discussed the concept formation and development status of regenerative rehabilitation, and the future development and potential leading value of regenerative rehabilitation field.
Collapse
Affiliation(s)
- J Wu
- Department of Burns and Plastic Surgery, Department of Wound Repair, the First Affiliated Hospital of Shenzhen University, Shenzhen 518035, China
| | - Y T Wei
- Department of Burns and Plastic Surgery, Department of Wound Repair, the First Affiliated Hospital of Shenzhen University, Shenzhen 518035, China
| |
Collapse
|
13
|
Wu W, Zhao Z, Wang Y, Zhu G, Tan K, Liu M, Li L. Biomechanical Effects of Mechanical Stress on Cells Involved in Fracture Healing. Orthop Surg 2024; 16:811-820. [PMID: 38439564 PMCID: PMC10984830 DOI: 10.1111/os.14026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 02/05/2024] [Accepted: 02/07/2024] [Indexed: 03/06/2024] Open
Abstract
Fracture healing is a complex staged repair process in which the mechanical environment plays a key role. Bone tissue is very sensitive to mechanical stress stimuli, and the literature suggests that appropriate stress can promote fracture healing by altering cellular function. However, fracture healing is a coupled process involving multiple cell types that balance and limit each other to ensure proper fracture healing. The main cells that function during different stages of fracture healing are different, and the types and molecular mechanisms of stress required are also different. Most previous studies have used a single mechanical stimulus on individual mechanosensitive cells, and there is no relatively uniform standard for the size and frequency of the mechanical stress. Analyzing the mechanisms underlying the effects of mechanical stimulation on the metabolic regulation of signaling pathways in cells such as in bone marrow mesenchymal stem cells (BMSCs), osteoblasts, chondrocytes, and osteoclasts is currently a challenging research hotspot. Grasping how stress affects the function of different cells at the molecular biology level can contribute to the refined management of fracture healing. Therefore, in this review, we summarize the relevant literature and describe the effects of mechanical stress on cells associated with fracture healing, and their possible signaling pathways, for the treatment of fractures and the further development of regenerative medicine.
Collapse
Affiliation(s)
- Weiyong Wu
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Zhihui Zhao
- Orthopedic Department, The Fourth Central Clinical School, Tianjin Medical University, Tianjin, China
| | - Yongqing Wang
- Orthopedic Department, The Fourth Central Clinical School, Tianjin Medical University, Tianjin, China
| | - Gengbao Zhu
- General Clinical Research Center, Anhui Wanbei Coal-Electricity Group General Hospital, Suzhou, China
| | - Kemeng Tan
- General Clinical Research Center, Anhui Wanbei Coal-Electricity Group General Hospital, Suzhou, China
| | - Meiyue Liu
- Orthopedic Department, The Fourth Central Clinical School, Tianjin Medical University, Tianjin, China
| | - Lili Li
- General Clinical Research Center, Anhui Wanbei Coal-Electricity Group General Hospital, Suzhou, China
| |
Collapse
|
14
|
Guo X, Lv M, Lin J, Guo J, Lin J, Li S, Sun Y, Zhang X. Latest Progress of LIPUS in Fracture Healing: A Mini-Review. JOURNAL OF ULTRASOUND IN MEDICINE : OFFICIAL JOURNAL OF THE AMERICAN INSTITUTE OF ULTRASOUND IN MEDICINE 2024; 43:643-655. [PMID: 38224522 DOI: 10.1002/jum.16403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 12/09/2023] [Accepted: 12/17/2023] [Indexed: 01/17/2024]
Abstract
The use of low-intensity pulsed ultrasound (LIPUS) for promoting fracture healing has been Food and Drug Administration (FDA)-approved since 1994 due to largely its non-thermal effects of sound flow sound radiation force and so on. Numerous clinical and animal studies have shown that LIPUS can accelerate the healing of fresh fractures, nonunions, and delayed unions in pulse mode regardless of LIPUS devices or circumstantial factors. Rare clinical studies show limitations of LIPUS for treating fractures with intramedullary nail fixation or low patient compliance. The biological effect is achieved by regulating various cellular behaviors involving mesenchymal stem/stromal cells (MSCs), osteoblasts, chondrocytes, and osteoclasts and with dose dependency on LIPUS intensity and time. Specifically, LIPUS promotes the osteogenic differentiation of MSCs through the ROCK-Cot/Tpl2-MEK-ERK signaling. Osteoblasts, in turn, respond to the mechanical signal of LIPUS through integrin, angiotensin type 1 (AT1), and PIEZO1 mechano-receptors, leading to the production of inflammatory factors such as COX-2, MCP-1, and MIP-1β fracture repair. LIPUS also induces CCN2 expression in chondrocytes thereby coordinating bone regeneration. Finally, LIPUS suppresses osteoclast differentiation and gene expression by interfering with the ERK/c-Fos/NFATc1 cascade. This mini-review revisits the known effects and mechanisms of LIPUS on bone fracture healing and strengthens the need for further investigation into the underlying mechanisms.
Collapse
Affiliation(s)
- Xin Guo
- School of Rehabilitation Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
- Department of Sports Medicine and Rehabilitation, Peking University Shenzhen Hospital, Shenzhen, China
| | - Maojiang Lv
- Department of Sports Medicine and Rehabilitation, Peking University Shenzhen Hospital, Shenzhen, China
- Zun Yi Medical University, Zhuhai, China
| | - Jie Lin
- Department of Joint Laboratory for Translational Medicine Research, Liaocheng People's Hospital, Liaocheng, China
| | - Jiang Guo
- Department of Sports Medicine and Rehabilitation, Peking University Shenzhen Hospital, Shenzhen, China
| | - Jianjing Lin
- Department of Sports Medicine and Rehabilitation, Peking University Shenzhen Hospital, Shenzhen, China
| | - Shun Li
- Department of Sports Medicine and Rehabilitation, Peking University Shenzhen Hospital, Shenzhen, China
| | - Yi Sun
- Department of Sports Medicine and Rehabilitation, Peking University Shenzhen Hospital, Shenzhen, China
- Department of Orthopaedics and Traumatology, The University of Hong Kong, Hong Kong SAR, China
| | - Xintao Zhang
- School of Rehabilitation Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
- Department of Sports Medicine and Rehabilitation, Peking University Shenzhen Hospital, Shenzhen, China
| |
Collapse
|
15
|
Bandyopadhyay A, Ghibhela B, Mandal BB. Current advances in engineering meniscal tissues: insights into 3D printing, injectable hydrogels and physical stimulation based strategies. Biofabrication 2024; 16:022006. [PMID: 38277686 DOI: 10.1088/1758-5090/ad22f0] [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: 09/15/2023] [Accepted: 01/26/2024] [Indexed: 01/28/2024]
Abstract
The knee meniscus is the cushioning fibro-cartilage tissue present in between the femoral condyles and tibial plateau of the knee joint. It is largely avascular in nature and suffers from a wide range of tears and injuries caused by accidents, trauma, active lifestyle of the populace and old age of individuals. Healing of the meniscus is especially difficult due to its avascularity and hence requires invasive arthroscopic approaches such as surgical resection, suturing or implantation. Though various tissue engineering approaches are proposed for the treatment of meniscus tears, three-dimensional (3D) printing/bioprinting, injectable hydrogels and physical stimulation involving modalities are gaining forefront in the past decade. A plethora of new printing approaches such as direct light photopolymerization and volumetric printing, injectable biomaterials loaded with growth factors and physical stimulation such as low-intensity ultrasound approaches are being added to the treatment portfolio along with the contemporary tear mitigation measures. This review discusses on the necessary design considerations, approaches for 3D modeling and design practices for meniscal tear treatments within the scope of tissue engineering and regeneration. Also, the suitable materials, cell sources, growth factors, fixation and lubrication strategies, mechanical stimulation approaches, 3D printing strategies and injectable hydrogels for meniscal tear management have been elaborated. We have also summarized potential technologies and the potential framework that could be the herald of the future of meniscus tissue engineering and repair approaches.
Collapse
Affiliation(s)
- Ashutosh Bandyopadhyay
- Biomaterials and Tissue Engineering Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, 781039, India
| | - Baishali Ghibhela
- Biomaterials and Tissue Engineering Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, 781039, India
| | - Biman B Mandal
- Biomaterials and Tissue Engineering Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, 781039, India
- Centre for Nanotechnology, Indian Institute of Technology Guwahati, Guwahati, Assam 781039, India
- Jyoti and Bhupat Mehta School of Health Sciences and Technology, Indian Institute of Technology Guwahati, Guwahati, Assam 781039, India
| |
Collapse
|
16
|
Zhong F, Cao S, Yang L, Liu J, Gui B, Wang H, Jiang N, Zhou Q, Deng Q. Low‑intensity pulsed ultrasound accelerates diabetic wound healing by ADSC‑derived exosomes via promoting the uptake of exosomes and enhancing angiogenesis. Int J Mol Med 2024; 53:23. [PMID: 38214291 PMCID: PMC10836517 DOI: 10.3892/ijmm.2024.5347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 11/15/2023] [Indexed: 01/13/2024] Open
Abstract
Diabetic wounds remain a great challenge for clinicians globally as a lack of effective radical treatment often results in poor prognosis. Exosomes derived from adipose‑derived stem cells (ADSC‑Exos) have been explored as an appealing nanodrug delivery system in the treatment of diabetic wounds. However, the short half‑life and low utilization efficiency of exosomes limit their therapeutic effects. Low‑intensity pulsed ultrasound (LIPUS) provides a non‑invasive mechanical stimulus to cells and exerts a number of biological effects such as cavitation and thermal effects. In the present study, whether LIPUS could enhance ADSC‑Exo‑mediated diabetic wound repair was investigated and its possible mechanism of action was explored. After isolation and characterization, ADSC‑Exos were injected into mice with diabetic wounds, then the mice were exposed to LIPUS irradiation. The control mice were subcutaneously injected with PBS. Wound healing assays, laser Doppler perfusion, Masson's staining and angiogenesis assays were used to assess treatment efficiency. Then, ADSC‑Exos were cocultured with human umbilical vein endothelial cells (HUVECs), and the proliferation, migration and tube formation of HUVECs were assessed. Moreover, the cellular uptake of ADSC‑Exos in vitro and in vivo was assessed to explore the synergistic mechanisms underlying the effects of LIPUS. The in vivo results demonstrated that LIPUS increased the uptake of exosomes and prolonged the residence of exosomes in the wound area, thus enhancing angiogenesis and accelerating wound repair in diabetic mice. The in vitro results further confirmed that LIPUS enhanced the uptake efficiency of ADSC‑Exos by 10.93‑fold and significantly increased the proliferation, migration and tubular formation of HUVECs. Therefore, the present study indicates that LIPUS is a promising strategy to improve the therapeutic effects of ADSC‑Exos in diabetic wounds by promoting the cellular uptake of exosomes and enhancing angiogenesis.
Collapse
Affiliation(s)
- Fanglu Zhong
- Department of Ultrasound, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Sheng Cao
- Department of Ultrasound, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Li Yang
- Department of Ultrasound, General Hospital of Central Theater Command, Wuhan, Hubei 430070, P.R. China
| | - Junbi Liu
- Department of Ultrasound, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Bin Gui
- Department of Ultrasound, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Hao Wang
- Department of Ultrasound, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Nan Jiang
- Department of Ultrasound, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Qing Zhou
- Department of Ultrasound, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Qing Deng
- Department of Ultrasound, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| |
Collapse
|
17
|
Wang W, Li Z, Yan Y, Wu S, Yao X, Gao C, Liu L, Yu Y. LIPUS-induced neurogenesis:A potential therapeutic strategy for cognitive dysfunction in traumatic brain injury. Exp Neurol 2024; 371:114588. [PMID: 37907126 DOI: 10.1016/j.expneurol.2023.114588] [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: 07/13/2023] [Revised: 10/02/2023] [Accepted: 10/25/2023] [Indexed: 11/02/2023]
Abstract
Traumatic brain injury (TBI) precipitates cellular membrane degeneration, phospholipid degradation, neuronal demise, impaired brain electrical activity, and compromised neuroplasticity, ultimately leading to acute and chronic brain dysfunction. Low-intensity pulsed ultrasound (LIPUS) is an emerging brain therapy with the characteristics of non-invasive, high spatial resolution, and high stimulation depth. Herein, we established a controlled cortical impact model to investigate the potential reparative mechanisms of LIPUS in TBI, employing a multi-faceted research methodology encompassing behavioral assessments, immunofluorescence, neuroelectrophysiology, scratch detection of primary cortical neurons, metabolomics and transcriptomics. Our findings demonstrate that LIPUS promotes hippocampal neurogenesis following brain injury, accomplished through the elevation of phosphatidylcholine levels in the hippocampus of TBI mice. Consequently, LIPUS enhances neural electrical activity and augments neural plasticity within the CA1 subregion of the hippocampus, effectively restoring neuronal function and cognitive capabilities in TBI mice. These findings shed light on the promising role of LIPUS in TBI brain rehabilitation, offering new perspectives and theoretical foundations for future studies in this domain.
Collapse
Affiliation(s)
- Wenzhu Wang
- China Rehabilitation Science Institute, China Rehabilitation Research Center, School of Rehabilitation, Capital Medical University, Beijing, PR China; Wenzhou Medical University, Zhejiang, PR China; Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, PR China; Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, PR China
| | - Zihan Li
- China Rehabilitation Science Institute, China Rehabilitation Research Center, School of Rehabilitation, Capital Medical University, Beijing, PR China; Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, PR China; Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, PR China
| | - Yitong Yan
- China Rehabilitation Science Institute, China Rehabilitation Research Center, School of Rehabilitation, Capital Medical University, Beijing, PR China; Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, PR China; Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, PR China
| | - Shuo Wu
- First Hospital of Qinhuangdao, Qinhuangdao, Hebei, PR China
| | - Xinyu Yao
- First Hospital of Qinhuangdao, Qinhuangdao, Hebei, PR China
| | - Chen Gao
- China Rehabilitation Science Institute, China Rehabilitation Research Center, School of Rehabilitation, Capital Medical University, Beijing, PR China; Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, PR China; Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, PR China
| | - Lanxiang Liu
- First Hospital of Qinhuangdao, Qinhuangdao, Hebei, PR China.
| | - Yan Yu
- China Rehabilitation Science Institute, China Rehabilitation Research Center, School of Rehabilitation, Capital Medical University, Beijing, PR China; Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, PR China; Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, PR China.
| |
Collapse
|
18
|
Zhu H, He M, Wang Y, Zhang Y, Dong J, Chen B, Li Y, Zhou L, Du L, Liu Y, Zhang W, Ta D, Duan S. Low-intensity pulsed ultrasound alleviates doxorubicin-induced cardiotoxicity via inhibition of S100a8/a9-mediated cardiac recruitment of neutrophils. Bioeng Transl Med 2023; 8:e10570. [PMID: 38023700 PMCID: PMC10658545 DOI: 10.1002/btm2.10570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 06/05/2023] [Accepted: 06/25/2023] [Indexed: 12/01/2023] Open
Abstract
Doxorubicin (DOX)-induced cardiotoxicity limits its broad use as a chemotherapy agent. The development of effective and non-invasive strategies to prevent DOX-associated adverse cardiac events is urgently needed. We aimed to examine whether and how low-intensity pulsed ultrasound (LIPUS) plays a protective role in DOX-induced cardiotoxicity. Male C57BL/6J mice were used to establish models of both acute and chronic DOX-induced cardiomyopathy. Non-invasive LIPUS therapy was conducted for four consecutive days after DOX administration. Cardiac contractile function was evaluated by echocardiography. Myocardial apoptosis, oxidative stress, and fibrosis were analyzed using terminal deoxynucleotidyl transferase-mediated dUTP nick end labelling (TUNEL) staining, dihydroethidium (DHE) staining, and picrosirius red staining assays. RNA-seq analysis was performed to unbiasedly explore the possible downstream regulatory mechanisms. Neutrophil recruitment and infiltration in the heart were analyzed by flow cytometry. The S100a8/a9 inhibitor ABR-238901 was utilized to identify the effect of S100a8/a9 signaling. We found that LIPUS therapy elicited a great benefit on DOX-induced heart contractile dysfunction in both acute and chronic DOX models. Chronic DOX administration increased serum creatine kinase and lactate dehydrogenase levels, as well as myocardial apoptosis, all of which were significantly mitigated by LIPUS. In addition, LIPUS treatment prevented chronic DOX-induced cardiac oxidative stress and fibrosis. RNA-seq analysis revealed that LIPUS treatment partially reversed alterations of gene expression induced by DOX. Gene ontology (GO) analysis of the downregulated genes between DOX-LIPUS and DOX-Sham groups indicated that inhibition of neutrophil chemotaxis might be involved in the protective effects of LIPUS therapy. Flow cytometry analysis illustrated the inhibitory effects of LIPUS on DOX-induced neutrophil recruitment and infiltration in the heart. Moreover, S100 calcium binding protein A8/A9 (S100a8/a9) was identified as a potential key target of LIPUS therapy. S100a8/a9 inhibition by ABR-238901 showed a similar heart protective effect against DOX-induced cardiomyopathy to LIPUS treatment. LIPUS therapy prevents DOX-induced cardiotoxicity through inhibition of S100a8/a9-mediated neutrophil recruitment to the heart, suggesting its potential application in cancer patients undergoing chemotherapy with DOX.
Collapse
Affiliation(s)
- Hong Zhu
- Laboratory of Oral Microbiota and Systemic DiseasesShanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of MedicineShanghaiChina
- Translational Medical Center for Stem Cell Therapy & Institutes for Regenerative Medicine, Shanghai East Hospital, Tongji University School of MedicineShanghaiChina
| | - Min He
- Center for Biomedical Engineering, School of Information Science and Technology, Fudan UniversityShanghaiChina
| | - Yong‐Li Wang
- Laboratory of Oral Microbiota and Systemic DiseasesShanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of MedicineShanghaiChina
- National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of StomatologyShanghaiChina
| | - Yuanxin Zhang
- Department of CardiologyNinth People's Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Jingsong Dong
- Center for Biomedical Engineering, School of Information Science and Technology, Fudan UniversityShanghaiChina
| | - Bo‐Yan Chen
- Laboratory of Oral Microbiota and Systemic DiseasesShanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of MedicineShanghaiChina
- National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of StomatologyShanghaiChina
| | - Yu‐Lin Li
- Laboratory of Oral Microbiota and Systemic DiseasesShanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of MedicineShanghaiChina
- National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of StomatologyShanghaiChina
| | - Lu‐Jun Zhou
- Laboratory of Oral Microbiota and Systemic DiseasesShanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of MedicineShanghaiChina
- National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of StomatologyShanghaiChina
| | - Lin‐Juan Du
- Laboratory of Oral Microbiota and Systemic DiseasesShanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of MedicineShanghaiChina
- National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of StomatologyShanghaiChina
| | - Yuan Liu
- Laboratory of Oral Microbiota and Systemic DiseasesShanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of MedicineShanghaiChina
- National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of StomatologyShanghaiChina
| | - Wu‐Chang Zhang
- Laboratory of Oral Microbiota and Systemic DiseasesShanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of MedicineShanghaiChina
- National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of StomatologyShanghaiChina
| | - Dean Ta
- Center for Biomedical Engineering, School of Information Science and Technology, Fudan UniversityShanghaiChina
- Department of Rehabilitation MedicineHuashan Hospital, Fudan UniversityShanghaiChina
| | - Sheng‐Zhong Duan
- Laboratory of Oral Microbiota and Systemic DiseasesShanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of MedicineShanghaiChina
- National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of StomatologyShanghaiChina
| |
Collapse
|
19
|
Jia W, Zhou Z, Zhan W. Musculoskeletal Biomaterials: Stimulated and Synergized with Low Intensity Pulsed Ultrasound. J Funct Biomater 2023; 14:504. [PMID: 37888169 PMCID: PMC10607075 DOI: 10.3390/jfb14100504] [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: 08/11/2023] [Revised: 09/10/2023] [Accepted: 09/21/2023] [Indexed: 10/28/2023] Open
Abstract
Clinical biophysical stimulating strategies, which have significant effects on improving the function of organs or treating diseases by causing the salutary response of body, have shown many advantages, such as non-invasiveness, few side effects, and controllable treatment process. As a critical technique for stimulation, the low intensity pulsed ultrasound (LIPUS) has been explored in regulating osteogenesis, which has presented great promise in bone repair by delivering a combined effect with biomaterials. This review summarizes the musculoskeletal biomaterials that can be synergized with LIPUS for enhanced biomedical application, including bone regeneration, spinal fusion, osteonecrosis/osteolysis, cartilage repair, and nerve regeneration. Different types of biomaterials are categorized for summary and evaluation. In each subtype, the verified biological mechanisms are listed in a table or graphs to prove how LIPUS was effective in improving musculoskeletal tissue regeneration. Meanwhile, the acoustic excitation parameters of LIPUS that were promising to be effective for further musculoskeletal tissue engineering are discussed, as well as their limitations and some perspectives for future research. Overall, coupled with biomimetic scaffolds and platforms, LIPUS may be a powerful therapeutic approach to accelerate musculoskeletal tissue repair and even in other regenerative medicine applications.
Collapse
Affiliation(s)
- Wanru Jia
- Department of Ultrasound, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China;
| | - Zifei Zhou
- Department of Orthopedics, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Weiwei Zhan
- Department of Ultrasound, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China;
| |
Collapse
|
20
|
Mohamad Yusoff F, Higashi Y. Mesenchymal Stem/Stromal Cells for Therapeutic Angiogenesis. Cells 2023; 12:2162. [PMID: 37681894 PMCID: PMC10486439 DOI: 10.3390/cells12172162] [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: 07/13/2023] [Revised: 08/18/2023] [Accepted: 08/25/2023] [Indexed: 09/09/2023] Open
Abstract
Mesenchymal stem/stromal cells (MSCs) are known to possess medicinal properties to facilitate vascular regeneration. Recent advances in the understanding of the utilities of MSCs in physiological/pathological tissue repair and technologies in isolation, expansion, and enhancement strategies have led to the use of MSCs for vascular disease-related treatments. Various conditions, including chronic arterial occlusive disease, diabetic ulcers, and chronic wounds, cause significant morbidity in patients. Therapeutic angiogenesis by cell therapy has led to the possibilities of treatment options in promoting angiogenesis, treating chronic wounds, and improving amputation-free survival. Current perspectives on the options for the use of MSCs for therapeutic angiogenesis in vascular research and in medicine, either as a monotherapy or in combination with conventional interventions, for treating patients with peripheral artery diseases are discussed in this review.
Collapse
Affiliation(s)
- Farina Mohamad Yusoff
- Department of Regenerative Medicine, Division of Radiation Medical Science, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima 734-8553, Japan;
| | - Yukihito Higashi
- Department of Regenerative Medicine, Division of Radiation Medical Science, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima 734-8553, Japan;
- Division of Regeneration and Medicine, Hiroshima University Hospital, Hiroshima 734-8551, Japan
| |
Collapse
|
21
|
Zhou J, Zhu Y, Ai D, Zhou M, Li H, Fu Y, Song J. Low-intensity pulsed ultrasound regulates osteoblast-osteoclast crosstalk via EphrinB2/EphB4 signaling for orthodontic alveolar bone remodeling. Front Bioeng Biotechnol 2023; 11:1192720. [PMID: 37425367 PMCID: PMC10326439 DOI: 10.3389/fbioe.2023.1192720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 06/16/2023] [Indexed: 07/11/2023] Open
Abstract
Background: The limited regenerative potential of periodontal tissue remains a challenge in orthodontic treatment, especially with respect to alveolar bone remodeling. The dynamic balance between the bone formation of osteoblasts and the bone resorption of osteoclasts controls bone homeostasis. The osteogenic effect of low-intensity pulsed ultrasound (LIPUS) is widely accepted, so LIPUS is expected to be a promising method for alveolar bone regeneration. Osteogenesis is regulated by the acoustic mechanical effect of LIPUS, while the cellular perception, transduction mode and response regulation mechanism of LIPUS stimuli are still unclear. This study aimed to explore the effects of LIPUS on osteogenesis by osteoblast-osteoclast crosstalk and the underlying regulation mechanism. Methods: The effects of LIPUS on orthodontic tooth movement (OTM) and alveolar bone remodeling were investigated via rat model by histomorphological analysis. Mouse bone marrow mesenchymal stem cells (BMSCs) and bone marrow monocytes (BMMs) were purified and used as BMSC-derived osteoblasts and BMM-derived osteoclasts, respectively. The osteoblast-osteoclast co-culture system was used to evaluate the effect of LIPUS on cell differentiation and intercellular crosstalk by Alkaline phosphatase (ALP), Alizarin Red S (ARS), tartrate-resistant acid phosphatase (TRAP) staining, real-time quantitative PCR, western blotting and immunofluorescence. Results: LIPUS was found to improve OTM and alveolar bone remodeling in vivo, promote differentiation and EphB4 expression in BMSC-derived osteoblasts in vitro, particularly when cells were directly co-cultured with BMM-derived osteoclasts. LIPUS enhanced EphrinB2/EphB4 interaction between osteoblasts and osteoclasts in alveolar bone, activated the EphB4 receptor on osteoblasts membrane, transduced LIPUS-related mechanical signals to the intracellular cytoskeleton, and gave rise to the nuclear translocation of YAP in Hippo signaling pathway, thus regulating cell migration and osteogenic differentiation. Conclusions: This study shows that LIPUS modulates bone homeostasis by osteoblast-osteoclast crosstalk via EphrinB2/EphB4 signaling, which benefits the balance between OTM and alveolar bone remodeling.
Collapse
Affiliation(s)
- Jie Zhou
- College of Stomatology, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Yanlin Zhu
- College of Stomatology, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Dongqing Ai
- College of Stomatology, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Mengjiao Zhou
- College of Stomatology, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Han Li
- College of Stomatology, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Yiru Fu
- College of Stomatology, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Jinlin Song
- College of Stomatology, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| |
Collapse
|
22
|
Ultrasound-targeted microbubble destruction remodels tumour microenvironment to improve immunotherapeutic effect. Br J Cancer 2023; 128:715-725. [PMID: 36463323 PMCID: PMC9977958 DOI: 10.1038/s41416-022-02076-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 11/10/2022] [Accepted: 11/15/2022] [Indexed: 12/04/2022] Open
Abstract
Cancer immunotherapy (CIT) has gained increasing attention and made promising progress in recent years, especially immune checkpoint inhibitors such as antibodies blocking programmed cell death 1/programmed cell death ligand 1 (PD-1/PD-L1) and cytotoxic T lymphocyte-associated protein 4 (CTLA-4). However, its therapeutic efficacy is only 10-30% in solid tumours and treatment sensitivity needs to be improved. The complex tissue environment in which cancers originate is known as the tumour microenvironment (TME) and the complicated and dynamic TME is correlated with the efficacy of immunotherapy. Ultrasound-targeted microbubble destruction (UTMD) is an emerging technology that integrates diagnosis and therapy, which has garnered much traction due to non-invasive, targeted drug delivery and gene transfection characteristics. UTMD has also been studied to remodel TME and improve the efficacy of CIT. In this review, we analyse the effects of UTMD on various components of TME, including CD8+ T cells, tumour-infiltrating myeloid cells, regulatory T cells, natural killer cells and tumour vasculature. Moreover, UTMD enhances the permeability of the blood-brain barrier to facilitate drug delivery, thus improving CIT efficacy in vivo animal experiments. Based on this, we highlight the potential of immunotherapy against various cancer species and the clinical application prospects of UTMD.
Collapse
|
23
|
Qin H, Du L, Luo Z, He Z, Wang Q, Chen S, Zhu YL. The therapeutic effects of low-intensity pulsed ultrasound in musculoskeletal soft tissue injuries: Focusing on the molecular mechanism. Front Bioeng Biotechnol 2022; 10:1080430. [PMID: 36588943 PMCID: PMC9800839 DOI: 10.3389/fbioe.2022.1080430] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 11/28/2022] [Indexed: 12/23/2022] Open
Abstract
Musculoskeletal soft tissue injuries are very common and usually occur during both sporting and everyday activities. The intervention of adjuvant therapies to promote tissue regeneration is of great importance to improving people's quality of life and extending their productive lives. Though many studies have focused on the positive results and effectiveness of the LIPUS on soft tissue, the molecular mechanisms standing behind LIPUS effects are much less explored and reported, especially the intracellular signaling pathways. We incorporated all research on LIPUS in soft tissue diseases since 2005 and summarized studies that uncovered the intracellular molecular mechanism. This review will also provide the latest evidence-based research progress in this field and suggest research directions for future experiments.
Collapse
Affiliation(s)
- Haocheng Qin
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Liang Du
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Zhiwen Luo
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Zhong He
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Qing Wang
- Department of Orthopedics, Kunshan Hospital of Chinese Medicine, Suzhou, China
| | - Shiyi Chen
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Yu-Lian Zhu
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
24
|
Matsuzaka Y, Yashiro R. Regulation of Extracellular Vesicle-Mediated Immune Responses against Antigen-Specific Presentation. Vaccines (Basel) 2022; 10:1691. [PMID: 36298556 PMCID: PMC9607341 DOI: 10.3390/vaccines10101691] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 09/28/2022] [Accepted: 10/04/2022] [Indexed: 11/24/2022] Open
Abstract
Extracellular vesicles (EVs) produced by various immune cells, including B and T cells, macrophages, dendritic cells (DCs), natural killer (NK) cells, and mast cells, mediate intercellular communication and have attracted much attention owing to the novel delivery system of molecules in vivo. DCs are among the most active exosome-secreting cells of the immune system. EVs produced by cancer cells contain cancer antigens; therefore, the development of vaccine therapy that does not require the identification of cancer antigens using cancer-cell-derived EVs may have significant clinical implications. In this review, we summarise the molecular mechanisms underlying EV-based immune responses and their therapeutic effects on tumour vaccination.
Collapse
Affiliation(s)
- Yasunari Matsuzaka
- Division of Molecular and Medical Genetics, Center for Gene and Cell Therapy, The Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo 108-8639, Japan
- Administrative Section of Radiation Protection, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira 187-8551, Tokyo, Japan
| | - Ryu Yashiro
- Administrative Section of Radiation Protection, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira 187-8551, Tokyo, Japan
- Department of Infectious Diseases, Kyorin University School of Medicine, 6-20-2 Shinkawa, Mitaka-shi 181-8611, Tokyo, Japan
| |
Collapse
|