1
|
Wen Z, Li S, Liu Y, Liu X, Qiu H, Che Y, Bian L, Zhou M. An engineered M2 macrophage-derived exosomes-loaded electrospun biomimetic periosteum promotes cell recruitment, immunoregulation, and angiogenesis in bone regeneration. Bioact Mater 2025; 50:95-115. [PMID: 40242509 PMCID: PMC12002949 DOI: 10.1016/j.bioactmat.2025.03.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 02/04/2025] [Accepted: 03/27/2025] [Indexed: 04/18/2025] Open
Abstract
The periosteum, a fibrous tissue membrane covering bone surfaces, is critical to osteogenesis and angiogenesis in bone reconstruction. Artificial periostea have been widely developed for bone defect repair, but most of these are lacking of periosteal bioactivity. Herein, a biomimetic periosteum (termed PEC-Apt-NP-Exo) is prepared based on an electrospun membrane combined with engineered exosomes (Exos). The electrospun membrane is fabricated using poly(ε-caprolactone) (core)-periosteal decellularized extracellular matrix (shell) fibers via coaxial electrospinning, to mimic the fibrous structure, mechanical property, and tissue microenvironment of natural periosteum. The engineered Exos derived from M2 macrophages are functionalized by surface modification of bone marrow mesenchymal stem cell (BMSC)-specific aptamers to further enhance cell recruitment, immunoregulation, and angiogenesis in bone healing. The engineered Exos are covalently bonded to the electrospun membrane, to achieve rich loading and long-term effects of Exos. In vitro experiments demonstrate that the biomimetic periosteum promotes BMSC migration and osteogenic differentiation via Rap1/PI3K/AKT signaling pathway, and enhances vascular endothelial growth factor secretion from BMSCs to facilitate angiogenesis. In vivo studies reveal that the biomimetic periosteum promotes new bone formation in large bone defect repair by inducing M2 macrophage polarization, endogenous BMSC recruitment, osteogenic differentiation, and vascularization. This research provides valuable insights into the development of a multifunctional biomimetic periosteum for bone regeneration.
Collapse
Affiliation(s)
- Zhuohao Wen
- Department of Stomatology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Shuyi Li
- Department of Stomatology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Yi Liu
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 511442, China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, China
- Guangdong Provincial Key Laboratory of Biomedical Engineering, South China University of Technology, Guangzhou, 510006, China
| | - Xueyan Liu
- Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, 510182, China
| | - Huiguo Qiu
- Zhuhai Stomatological Hospital, Zhuhai, 519000, China
| | - Yuejuan Che
- Department of Biomedical Engineering, School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510641, China
- Department of Anesthesiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
| | - Liming Bian
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 511442, China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, China
- Guangdong Provincial Key Laboratory of Biomedical Engineering, South China University of Technology, Guangzhou, 510006, China
| | - Miao Zhou
- Department of Stomatology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| |
Collapse
|
2
|
Wang Y, Pan Z, Wang Q, Shu Y, Tan Z, Chen Y, He J, Wang J, Wang J, Yan J. Sequential SDF-1/CGRP-releasing smart composite hydrogel promotes osteoporotic fracture healing by targeting sensory nerve-regulated bone remodeling. Mater Today Bio 2025; 32:101750. [PMID: 40331153 PMCID: PMC12054128 DOI: 10.1016/j.mtbio.2025.101750] [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: 11/24/2024] [Revised: 04/04/2025] [Accepted: 04/09/2025] [Indexed: 05/08/2025] Open
Abstract
Osteoporotic fractures typically exhibit delayed healing due to impaired cell recruitment, chronic inflammation, and disrupted neurovascular signaling. Sensory nerve signaling plays a crucial role in fracture repair, and its deficiency is a significant factor leading to delayed healing. Addressing these deficiencies is crucial to overcoming the challenges associated with delayed bone repair in osteoporosis. In this study, a smart composite hydrogel (denoted as OCS-MPC) was synthesized by embedding CGRP-functionalized polydopamine-coated MXene nanosheets (MXene/PDA/CGRP) into boronic acid-modified oxidized hyaluronic acid-crosslinked carboxymethyl chitosan (OHA-PBA/CMCS) hydrogel loaded with SDF-1. OCS-MPC hydrogel enables the controlled release of SDF-1 and CGRP, aiming to promote early callus formation and late-stage callus remodeling in osteoporotic fractures. Due to dynamic crosslinking via imine and borate ester bonds, OCS-MPC exhibits rapid gelation, injectability, and self-healing properties. In vitro experiments demonstrated excellent osteogenic, angiogenic, and neurogenic properties of OCS-MPC hydrogel. In vivo studies using an osteoporotic femoral fracture model showed that OCS-MPC hydrogel enhanced MSCs recruitment via the SDF-1/CXCR4 signaling axis, significantly improving callus formation in the early stages of fracture repair. Additionally, OCS-MPC hydrogel significantly promoted callus mineralization and remodeling in the later stages of osteoporotic fracture healing through enhancing CGRP signaling. Immunofluorescence analysis further confirmed increased expression of TUBB3, CGRP, and CD31, indicating successful regeneration of the neurovascular network. These findings highlight the potential of OCS-MPC hydrogel in addressing both early and late-stage challenges of osteoporotic fracture healing, providing a promising therapeutic strategy for enhancing bone regeneration in osteoporotic patients.
Collapse
Affiliation(s)
- Yuan Wang
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
- Department of Orthopedics, TongRen Hospital, School of Medicine Shanghai Jiao Tong University, Shanghai, 200336, China
| | - Zhen Pan
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200235, China
| | - Qianliang Wang
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
| | - Yuexia Shu
- Department of Orthopedics, TongRen Hospital, School of Medicine Shanghai Jiao Tong University, Shanghai, 200336, China
- Laboratory of Key Technology and Materials in Minimally Invasive Spine Surgery, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200336, China
- Center for Spinal Minimally Invasive Research, Shanghai Jiao Tong University, Shanghai, 200336, China
| | - Zhenyu Tan
- Department of Pathology, Tongji Hospital, Tongji University, Shanghai, 200065, China
| | - Yujie Chen
- Department of Orthopedics, TongRen Hospital, School of Medicine Shanghai Jiao Tong University, Shanghai, 200336, China
- Laboratory of Key Technology and Materials in Minimally Invasive Spine Surgery, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200336, China
- Center for Spinal Minimally Invasive Research, Shanghai Jiao Tong University, Shanghai, 200336, China
| | - Jieming He
- Department of Orthopedics, TongRen Hospital, School of Medicine Shanghai Jiao Tong University, Shanghai, 200336, China
| | - Jia Wang
- Department of Orthopedics, TongRen Hospital, School of Medicine Shanghai Jiao Tong University, Shanghai, 200336, China
| | - Jielin Wang
- Department of Orthopedics, TongRen Hospital, School of Medicine Shanghai Jiao Tong University, Shanghai, 200336, China
- Laboratory of Key Technology and Materials in Minimally Invasive Spine Surgery, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200336, China
- Center for Spinal Minimally Invasive Research, Shanghai Jiao Tong University, Shanghai, 200336, China
| | - Jun Yan
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
| |
Collapse
|
3
|
Yang L, Xu Z, Liu J, Chang X, Ren Z, Xiao W. Multi-omics insights into bone tissue injury and healing: bridging orthopedic trauma and regenerative medicine. BURNS & TRAUMA 2025; 13:tkaf019. [PMID: 40438296 PMCID: PMC12118463 DOI: 10.1093/burnst/tkaf019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Revised: 06/18/2024] [Accepted: 02/27/2025] [Indexed: 06/01/2025]
Abstract
To preserve functionality, bone is an active tissue that can constantly reconstruct itself through modeling and remodeling. It plays critical roles in the body, including maintaining mineral homeostasis, serving as the adult human body's core site of hematopoiesis, and supporting the structures of the body's soft tissues. It possesses the natural regeneration capacity, but large and complex lesions often require surgical intervention. Multiple omics integrate proteomics, metabolomics, genomics, and transcriptomics to provide a comprehensive understanding of biological processes like bone tissue injury and healing in bone tissue regeneration and engineering. Recently, bone tissue engineering and regenerative medicines have offered promising tools for bone regeneration using a multi-omics approach. Thus, this article will highlight the role of multiple omics in understanding bone tissue injury and healing. It will discuss the role of bone tissue engineering in developing bone substitutes that can replace translational medicine. Lastly, new developments in bone tissue engineering and regenerative medicine, along with multi-omics approaches, offer promising tools for bone regeneration.
Collapse
Affiliation(s)
- Liyu Yang
- Department of Orthopedics, Shengjing Hospital of China Medical University, 36 Sanhao Street, Heping District, Shenyang, Liaoning 110004, China
| | - Zhijie Xu
- Department of Orthopedics, Shengjing Hospital of China Medical University, 36 Sanhao Street, Heping District, Shenyang, Liaoning 110004, China
| | - Jie Liu
- Department of Orthopedics, Shengjing Hospital of China Medical University, 36 Sanhao Street, Heping District, Shenyang, Liaoning 110004, China
- Department of Epidemiology, School of Public Health, China Medical University, 77 Puhe Road, Shenbei New District, Shenyang, Liaoning 110013, China
| | - Xiyue Chang
- Department of Orthopedics, Shengjing Hospital of China Medical University, 36 Sanhao Street, Heping District, Shenyang, Liaoning 110004, China
- Department of Epidemiology, School of Public Health, China Medical University, 77 Puhe Road, Shenbei New District, Shenyang, Liaoning 110013, China
| | - Zhaozhou Ren
- Department of Orthopedics, Shengjing Hospital of China Medical University, 36 Sanhao Street, Heping District, Shenyang, Liaoning 110004, China
| | - Wan’an Xiao
- Department of Orthopedics, Shengjing Hospital of China Medical University, 36 Sanhao Street, Heping District, Shenyang, Liaoning 110004, China
| |
Collapse
|
4
|
Han J, Li Z, Du J, Zhang Q, Ge S, Liu H, Ma B. Natural collagen scaffold with intrinsic piezoelectricity for enhanced bone regeneration. Mater Today Bio 2025; 31:101532. [PMID: 39968523 PMCID: PMC11834078 DOI: 10.1016/j.mtbio.2025.101532] [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: 11/25/2024] [Revised: 01/27/2025] [Accepted: 01/28/2025] [Indexed: 02/20/2025] Open
Abstract
Materials-mediated piezoelectric signals have been widely applied in bone regeneration. Collagen is the most abundant protein in the human body, and native collagen with complete tertiary structure shows efficient piezoelectricity. However, the traditional collagen scaffolds are lack of piezoelectricity due to the destruction of the complete tertiary structure. Here, natural collagen scaffolds with the complete tertiary structure were prepared. Alkali treatment made the collagen scaffold lose piezoelectricity. The collagen with/without piezoelectricity (PiezoCol/NCol) scaffolds both possessed good cytocompatibility and promoted cell adhesion. After being implanted subcutaneously, the NCol scaffold almost did not affect bone regeneration with/without ultrasound treatment. However, under ultrasound treatment, the PiezoCol scaffold promoted the new bone formation with enhanced osteogenic differentiation, angiogenesis, and neural differentiation, meaning that piezoelectricity endows collagen with satisfactory promotion for bone regeneration. Meanwhile, the PiezoCol scaffold can also accelerate bone formation without ultrasound treatment, which should be attributed to the daily exercise-caused weak piezoelectric stimulation. Further, the proteomic analysis revealed the mechanism by which the PiezoCol scaffold promoted bone tissue formation via mainly upregulating the PI3K-Akt signaling pathway. This study provides a new strategy to enhance the osteoinduction of collagen scaffold for bone regeneration by maintaining intrinsic piezoelectricity.
Collapse
Affiliation(s)
- Jing Han
- Department of Tissue Engineering & Periodontology, School and Hospital of Stomatology, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, Shandong, 250012, China
| | - Zhao Li
- College of Materials Science and Engineering, Qingdao University of Science & Technology, Qingdao, Shandong, 266061, China
| | - Jing Du
- Department of Tissue Engineering & Periodontology, School and Hospital of Stomatology, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, Shandong, 250012, China
| | - Qun Zhang
- Department of Tissue Engineering & Periodontology, School and Hospital of Stomatology, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, Shandong, 250012, China
| | - Shaohua Ge
- Department of Tissue Engineering & Periodontology, School and Hospital of Stomatology, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, Shandong, 250012, China
| | - Hong Liu
- State Key Laboratory of Crystal Materials, Shandong University, Jinan, Shandong, 250013, China
| | - Baojin Ma
- Department of Tissue Engineering & Periodontology, School and Hospital of Stomatology, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, Shandong, 250012, China
| |
Collapse
|
5
|
Ai S, Xie Z, Li N, Zhao R, Qu X, Zhou H, Tang D, Zhang J, Luo X. Bibliometric and visualized analysis of the applications of exosomes for bone regeneration. Front Cell Dev Biol 2025; 13:1552727. [PMID: 40166633 PMCID: PMC11955700 DOI: 10.3389/fcell.2025.1552727] [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: 12/29/2024] [Accepted: 02/20/2025] [Indexed: 04/02/2025] Open
Abstract
Background Bone defect, a common orthopedic condition, is characterized by a lengthy and impactful treatment period, posing a considerable challenge in clinical settings. Medical technology has advanced notably, and has effectively treated an increasing number of patients with bone defects. Consequently, there has been an explosion of research articles on bone regeneration, including a substantial number on the application of exosomes. Exosomes, especially those derived from stem cells, have been confirmed to be effective in bone regeneration and have garnered widespread attention in the last decade. Therefore, this study conducted a bibliometric analysis on publications related to the application of exosomes for bone regeneration. The objectives are to explore the development history and research hotspots in this field over the past 10 years, predict future development trends, and provide guidance for subsequent research. Methods The Web of Science Core Collection (WoSCC) database was searched for articles related to exosomes and bone regeneration published from 1 January 2014, to 31 December 2023. The collected literature was analyzed using software such as Microsoft Excel, CiteSpace 6.3R1, VOSviewer 1.6.20, and the bibliometric online platform (https://bibliometric.com). Results A total of 3,004 articles published by 2,729 institutions from 68 countries were included in this study. The number of articles on the application of exosomes for bone regeneration has increased annually over the last decade. China was the most prolific country in this field, with a total of 1,468 papers; Shanghai Jiao Tong University (China) was the institution with the highest number of publications (117 publications). In terms of authors, Xin Wang, Yi Zhang, and Yang Wang were the three who published the highest number of papers, with 14 papers each. Co-citation analysis revealed that the article published by Valadi H in 2007 has the highest number of co-citations (270 times of quotation). Additionally, most research hotspots focused on the function of exosomes and the mechanism of action. Furthermore, the importance of osteoblast differentiation and angiogenesis in bone regeneration has also garnered significant attention from scholars in this field. Conclusion This study reviewed the research achievements on the application of exosomes for bone regeneration over the past 10 years, utilizing bibliometric analysis tools. It visualized the countries, institutions, authors, and journals that have made significant contributions to this field, revealed current research hotspots, and finally explored future development trends.
Collapse
Affiliation(s)
- Shuai Ai
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing Municipal Health Commission Key Laboratory of Musculoskeletal Regeneration and Translational Medicine, Orthopaedic Research Laboratory of Chongqing Medical University, Chongqing, China
| | - Zhou Xie
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing Municipal Health Commission Key Laboratory of Musculoskeletal Regeneration and Translational Medicine, Orthopaedic Research Laboratory of Chongqing Medical University, Chongqing, China
| | - Ningdao Li
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing Municipal Health Commission Key Laboratory of Musculoskeletal Regeneration and Translational Medicine, Orthopaedic Research Laboratory of Chongqing Medical University, Chongqing, China
| | - Runhan Zhao
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing Municipal Health Commission Key Laboratory of Musculoskeletal Regeneration and Translational Medicine, Orthopaedic Research Laboratory of Chongqing Medical University, Chongqing, China
| | - Xiao Qu
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing Municipal Health Commission Key Laboratory of Musculoskeletal Regeneration and Translational Medicine, Orthopaedic Research Laboratory of Chongqing Medical University, Chongqing, China
| | - Haining Zhou
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing Municipal Health Commission Key Laboratory of Musculoskeletal Regeneration and Translational Medicine, Orthopaedic Research Laboratory of Chongqing Medical University, Chongqing, China
| | - Dagang Tang
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing Municipal Health Commission Key Laboratory of Musculoskeletal Regeneration and Translational Medicine, Orthopaedic Research Laboratory of Chongqing Medical University, Chongqing, China
| | - Jun Zhang
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing Municipal Health Commission Key Laboratory of Musculoskeletal Regeneration and Translational Medicine, Orthopaedic Research Laboratory of Chongqing Medical University, Chongqing, China
| | - Xiaoji Luo
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing Municipal Health Commission Key Laboratory of Musculoskeletal Regeneration and Translational Medicine, Orthopaedic Research Laboratory of Chongqing Medical University, Chongqing, China
- The First Affiliated Hospital of Chongqing Medical and Pharmaceutical College, Chongqing, China
| |
Collapse
|
6
|
Liang W, Zhou C, Liu X, Xie Q, Xia L, Li Q, Lin H, Xiong X, Zhang H, Zheng Z, Zhao J. Synthetic Nanopillars for Stimulating Osteoblast Activity and Osteointegration in Bone-Related Disorders. Int J Nanomedicine 2025; 20:2205-2223. [PMID: 39990287 PMCID: PMC11847438 DOI: 10.2147/ijn.s501963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Accepted: 02/07/2025] [Indexed: 02/25/2025] Open
Abstract
Osteoporosis, osteoarthritis, and fractures are bone-related disorders that have a huge impact on the quality of life and healthcare systems worldwide. Traditional treatments, including bone grafts, have their limitations, with bone grafts often being rejected by the immune system and infected, making new treatments necessary. Nanopillars based on synthetic polymers have been demonstrated to be promising tools for bone regeneration and repair, showing to emulate the extracellular matrix composition, stimulate osteoblast activity and induce osteointegration. In this review, nanopillars fabrication techniques, such as electrospinning, nanoimprint lithography and self-assembly, also the state of the art of nanopillars technology are presented. Their role in modulating cellular responses via both physical and biochemical means, to enhance mineralization and to stabilize implants is also discussed. Additionally, their applications in treating bone-related disorders, eg promotion of fracture healing, augmentation of dental or orthopedic implants, and improvement of bone tissue engineering are discussed in the review. Using these focuses, each section examines opportunities and challenges (eg optimizing fabrication processes, improving biocompatibility, and investigating the integration of nanopillars with upcoming therapies like gene and stem cell therapy) for the potential of nanopillar technology. Finally, this review points out the requirement of scalable fabrication techniques, long term biocompatibility studies and multifunctional therapeutic strategies to fully employ the therapeutic applications of nanopillars in clinical scenarios. This review seeks to consolidate current knowledge of synthetic polymer based nanopillars and identify future directions for their use in bone related disorders through a comprehensive synthetic polymer nanopillar review.
Collapse
Affiliation(s)
- Wenqing Liang
- Department of Orthopedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, 316000, People’s Republic of China
| | - Chao Zhou
- Department of Orthopedics, Zhoushan Guanghua Hospital, Zhoushan, 316000, People’s Republic of China
| | - Xiankun Liu
- Department of Orthopedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, 316000, People’s Republic of China
| | - Qiong Xie
- Medical Research Center, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, 316000, People’s Republic of China
| | - Linying Xia
- Medical Research Center, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, 316000, People’s Republic of China
| | - Qingping Li
- Medical Research Center, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, 316000, People’s Republic of China
| | - Hongming Lin
- Department of Orthopedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, 316000, People’s Republic of China
| | - Xiaochun Xiong
- Department of Orthopedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, 316000, People’s Republic of China
| | - Hao Zhang
- Department of Orthopedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, 316000, People’s Republic of China
| | - Zeping Zheng
- Department of Orthopedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, 316000, People’s Republic of China
| | - Jiayi Zhao
- Department of Orthopedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, 316000, People’s Republic of China
| |
Collapse
|
7
|
Khoshbin E, Karkehabadi H, Salehi R, Farmany A, Najafi R, Abbasi R. Comparative study of nanohydroxyapatite-emdogain effects on apical papilla stem cell survival and differentiation. Biotechnol Lett 2025; 47:24. [PMID: 39907710 DOI: 10.1007/s10529-024-03557-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 09/26/2024] [Accepted: 12/16/2024] [Indexed: 02/06/2025]
Abstract
BACKGROUND The study was designed to explore the enhanced impact of nano-hydroxyapatite and emdogain on the survival and osteogenic/odontogenic differentiation of human stem cells isolated from the apical papilla (hSCAPs). MATERIALS AND METHODS In this in vitro trial, hSCAPS obtained from intact impacted immature third molars were confirmed to have characteristic cell surface markers, then exposed to nanohydroxyapatite, emdogain, and nanohydroxyapatite coated with emdogain for durations of 1-3 days. The survival of apical papilla stem cells was measured using a methyl thiazolyl tetrazolium assay. The quantitative reverse transcription polymerase chain reaction, alkaline phosphatase activity (ALP) and Alizarin red staining were used to evaluate osteogenic-odontogenic differentiation. Analysis of the data was done using one-way ANOVA, t-test, and Mann-Whitney test (α = 0.05). RESULTS At 1-3 days, emdogain exhibited no significant impact on the survival of human stem cells from the apical papilla. In contrast, nanohydroxyapatite (α > 0.05) and nanohydroxyapatite coated with emdogain demonstrated a notable reduction in cell survival compared to the control group (α < 0.05). The expression of dentin sialophosphoprotein, dentin matrix protein 1, and bone sialoprotein genes demonstrated a notable increase in the group treated with nanohydroxyapatite coated with emdogain compared to the other groups (α < 0.05), and furthermore, this group exhibited more pronounced mineralized nodules than the other experimental groups. CONCLUSION In contrast to nanohydroxyapatite, Emdogain did not demonstrate a detrimental effect on the survival of hSCAPs. Nanohydroxyapatite, emdogain, and nanohydroxyapatite coated with emdogain increased osteogenic/odontogenic differentiation of hSCAPs.
Collapse
Affiliation(s)
- Elham Khoshbin
- Department of Endodontics, School of Dentistry, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Hamed Karkehabadi
- Department of Endodontics, Dental Research Center, Avicenna Institute of Clinical Sciences, Avicenna Health Research Institute, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Razieh Salehi
- Department of Endodontics, School of Dentistry, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Abbas Farmany
- Dental Research Center, School of Dentistry, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Rezvan Najafi
- Department of Medical Molecular & Genetics, Faculty of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Roshanak Abbasi
- Department of Endodontics, School of Dentistry, Lorestan University of Medical Sciences, Khorramabad, Iran.
| |
Collapse
|
8
|
Sheikhi K, Ghaderi S, Firouzi H, Rahimibarghani S, Shabani E, Afkhami H, Yarahmadi A. Recent advances in mesenchymal stem cell therapy for multiple sclerosis: clinical applications and challenges. Front Cell Dev Biol 2025; 13:1517369. [PMID: 39963155 PMCID: PMC11830822 DOI: 10.3389/fcell.2025.1517369] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Accepted: 01/09/2025] [Indexed: 02/20/2025] Open
Abstract
Multiple sclerosis (MS), a chronic autoimmune disorder of the central nervous system (CNS), is characterized by inflammation, demyelination, and neurodegeneration, leading to diverse clinical manifestations such as fatigue, sensory impairment, and cognitive dysfunction. Current pharmacological treatments primarily target immune modulation but fail to arrest disease progression or entirely reverse CNS damage. Mesenchymal stem cell (MSC) therapy offers a promising alternative, leveraging its immunomodulatory, neuroprotective, and regenerative capabilities. This review provides an in-depth analysis of MSC mechanisms of action, including immune system regulation, promotion of remyelination, and neuroregeneration. It examines preclinical studies and clinical trials evaluating the efficacy, safety, and limitations of MSC therapy in various MS phenotypes. Special attention is given to challenges such as delivery routes, dosing regimens, and integrating MSCs with conventional therapies. By highlighting advancements and ongoing challenges, this review underscores the potential of MSCs to revolutionize MS treatment, paving the way for personalized and combinatory therapeutic approaches.
Collapse
Affiliation(s)
- Kamran Sheikhi
- Kurdistan University of Medical Sciences, Kurdistan, Iran
| | | | - Hassan Firouzi
- Department of Medical Laboratory, Faculty of Medicine, Sari Branch, Islamic Azad University, Sari, Iran
| | - Sarvenaz Rahimibarghani
- Department of Physical Medicine and Rehabilitation, Tehran University of Medical Sciences, Tehran, Iran
| | - Ehsan Shabani
- Department of Clinical Pharmacy, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, 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
| | - Aref Yarahmadi
- Department of Biology, Khorramabad Branch, Islamic Azad University, Khorramabad, Iran
| |
Collapse
|
9
|
Wang Y, Li Z, Yu R, Chen Y, Wang D, Zhao W, Ge S, Liu H, Li J. Metal-phenolic network biointerface-mediated cell regulation for bone tissue regeneration. Mater Today Bio 2025; 30:101400. [PMID: 39759849 PMCID: PMC11699301 DOI: 10.1016/j.mtbio.2024.101400] [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: 11/07/2024] [Revised: 12/07/2024] [Accepted: 12/09/2024] [Indexed: 01/07/2025] Open
Abstract
Bone tissue regeneration presents a significant challenge in clinical treatment due to inadequate coordination between implant materials and reparative cells at the biomaterial-bone interfaces. This gap underscores the necessity of enhancing interaction modulation between cells and biomaterials, which is a crucial focus in bone tissue engineering. Metal-polyphenolic networks (MPN) are novel inorganic-organic hybrid complexes that are formed through coordination interactions between phenolic ligands and metal ions. These networks provide a multifunctional platform for biomedical applications, with the potential for tailored design and modifications. Despite advances in understanding MPN and their role in bone tissue regeneration, a comprehensive overview of the related mechanisms is lacking. Here, we address this gap by focusing on MPN biointerface-mediated cellular regulatory mechanisms during bone regeneration. We begin by reviewing the natural healing processes of bone defects, followed by a detailed examination of MPN, including their constituents and distinctive characteristics. We then explore the regulatory influence of MPN biointerfaces on key cellular activities during bone regeneration. Additionally, we illustrate their primary applications in addressing inflammatory bone loss, regenerating critical-size bone defects, and enhancing implant-bone integration. In conclusion, this review elucidates how MPN-based interfaces facilitate effective bone tissue regeneration, advancing our understanding of material interface-mediated cellular control and the broader field of tissue engineering.
Collapse
Affiliation(s)
- Ying Wang
- Department of Biomaterials, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, 250012, China
| | - Zhibang Li
- Department of Biomaterials, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, 250012, China
| | - Ruiqing Yu
- Department of Biomaterials, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, 250012, China
| | - Yi Chen
- Department of Biomaterials, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, 250012, China
| | - Danyang Wang
- Department of Biomaterials, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, 250012, China
| | - Weiwei Zhao
- Department of Biomaterials, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, 250012, China
| | - Shaohua Ge
- Department of Biomaterials, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, 250012, China
| | - Hong Liu
- State Key Laboratory of Crystal Materials, Shandong University, Jinan, Shandong, 250100, China
| | - Jianhua Li
- Department of Biomaterials, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, 250012, China
| |
Collapse
|
10
|
Liu L, Liu W, Han Z, Shan Y, Xie Y, Wang J, Qi H, Xu Q. Extracellular Vesicles-in-Hydrogel (EViH) targeting pathophysiology for tissue repair. Bioact Mater 2025; 44:283-318. [PMID: 39507371 PMCID: PMC11539077 DOI: 10.1016/j.bioactmat.2024.10.017] [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: 04/06/2024] [Revised: 10/08/2024] [Accepted: 10/17/2024] [Indexed: 11/08/2024] Open
Abstract
Regenerative medicine endeavors to restore damaged tissues and organs utilizing biological approaches. Utilizing biomaterials to target and regulate the pathophysiological processes of injured tissues stands as a crucial method in propelling this field forward. The Extracellular Vesicles-in-Hydrogel (EViH) system amalgamates the advantages of extracellular vesicles (EVs) and hydrogels, rendering it a prominent biomaterial in regenerative medicine with substantial potential for clinical translation. This review elucidates the development and benefits of the EViH system in tissue regeneration, emphasizing the interaction and impact of EVs and hydrogels. Furthermore, it succinctly outlines the pathophysiological characteristics of various types of tissue injuries such as wounds, bone and cartilage injuries, cardiovascular diseases, nerve injuries, as well as liver and kidney injuries, underscoring how EViH systems target these processes to address related tissue damage. Lastly, it explores the challenges and prospects in further advancing EViH-based tissue regeneration, aiming to impart a comprehensive understanding of EViH. The objective is to furnish a thorough overview of EViH in enhancing regenerative medicine applications and to inspire researchers to devise innovative tissue engineering materials for regenerative medicine.
Collapse
Affiliation(s)
- Lubin Liu
- Department of Stomatology, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
- School of Stomatology, Qingdao University, Qingdao, 266023, China
| | - Wei Liu
- Department of Emergency Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266003, China
| | - Zeyu Han
- Department of Stomatology, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
- School of Stomatology, Qingdao University, Qingdao, 266023, China
| | - Yansheng Shan
- School of Stomatology, Qingdao University, Qingdao, 266023, China
| | - Yutong Xie
- Department of Stomatology, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
- School of Stomatology, Qingdao University, Qingdao, 266023, China
| | - Jialu Wang
- Department of Stomatology, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
- School of Stomatology, Qingdao University, Qingdao, 266023, China
| | - Hongzhao Qi
- Institute of Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China
| | - Quanchen Xu
- Department of Stomatology, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
- School of Stomatology, Qingdao University, Qingdao, 266023, China
| |
Collapse
|
11
|
Shanmugavadivu A, Selvamurugan N. Surface engineering of 3D-printed polylactic acid scaffolds with polydopamine and 4-methoxycinnamic acid-chitosan nanoparticles for bone regeneration. NANOSCALE ADVANCES 2025:d4na00768a. [PMID: 39886612 PMCID: PMC11776148 DOI: 10.1039/d4na00768a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Accepted: 01/17/2025] [Indexed: 02/01/2025]
Abstract
Bone remodeling, a continuous process of resorption and formation, is essential for maintaining skeletal integrity and mineral balance. However, in cases of critical bone defects where the natural bone remodeling capacity is insufficient, medical intervention is necessary. Traditional bone grafts have limitations such as donor site morbidity and availability, driving the search for bioengineered scaffold alternatives. The choice of biomaterial is crucial in scaffold design, as it provides a substrate that supports cell adhesion, proliferation, and differentiation. Poly-lactic acid (PLA) is known for its biocompatibility and biodegradability, but its hydrophobicity hinders cell attachment and tissue regeneration. To enhance PLA's bioactivity, we fabricated 3D-printed PLA scaffolds using fused deposition modelling. They were then surface-treated with NaOH to increase their reactivity, followed by polydopamine (PDA) and 4-methoxycinnamic acid (MCA)-loaded chitosan nanoparticle (nCS) coatings though polyelectrolyte complexation. Even though MCA, a polyphenolic, is known for its therapeutic properties, its osteogenic potential is not yet known. MCA treatment in mouse mesenchymal stem cells (mMSCs) promoted increased levels of Runx2 mRNA, a key bone transcription factor. Due to MCA's hydrophobic nature, nCS were used as carriers. The PLA/PDA/nCS-MCA scaffolds exhibited exceptional compressive strength and bioactivity. Biocompatibility tests confirmed that these scaffolds were non-cytotoxic to mMSCs. Overall, this study highlights the osteogenic potential of MCA and demonstrates the improved biocompatibility, bioactivity, wettability, and cell adhesion properties of the PDA/nCS-MCA-coated PLA scaffolds, positioning it as a promising material for bone tissue regeneration.
Collapse
Affiliation(s)
- Abinaya Shanmugavadivu
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology Kattankulathur Tamil Nadu 603203 India
| | - Nagarajan Selvamurugan
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology Kattankulathur Tamil Nadu 603203 India
| |
Collapse
|
12
|
Parmentier L, D'Haese S, Van der Meeren L, Szabó A, Skirtach AG, Dmitriev RI, Locs J, Van Vlierberghe S. Mimicking the Bone Extracellular Matrix through a Calcium Phosphate-Containing Thiol-Ene Cross-Linked Gelatin Composite. Biomacromolecules 2025; 26:332-340. [PMID: 39680045 DOI: 10.1021/acs.biomac.4c01182] [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: 12/17/2024]
Abstract
Hydroxyapatite (HAP) and amorphous calcium phosphate (ACP) nanoparticles were incorporated into a thiol-ene clickable gelatin network to elucidate to what extent osteogenic differentiation of human dental pulp- and adipose-derived stem cells (HDPSCs/HASCs) could be further boosted. ACP nanoparticles increased the specific surface area by 23% and reduced the density by 13% while maintaining a comparable particle size (ACP: 25 ± 3 nm; HAP: 27 ± 3 nm). Overall, the incorporation of ceramic nanoparticles did not significantly alter the mechanical properties of the ceramic-containing composites compared to the unsubstituted thiol-ene network. ACP nanoparticles at high concentrations promoted a 21-day osteogenic response in HASCs (72.09 ± 20.20 ng Ca2+/ng DNA) comparable to HDPSCs, with the latter showing high calcium production irrespective of the ceramic content (78.45 ± 10.87 ng Ca2+/ng DNA), suggesting that the provided cues must be optimized according to the investigated cell type toward a cell-interactive coating application stimulating osteogenesis.
Collapse
Affiliation(s)
- Laurens Parmentier
- Polymer Chemistry and Biomaterials (PBM) Group, Centre of Macromolecular Chemistry (CMaC), Department of Organic and Macromolecular Chemistry, Faculty of Sciences, Ghent University, Krijgslaan 281, Building S4, Ghent 9000, Belgium
| | - Sophie D'Haese
- Polymer Chemistry and Biomaterials (PBM) Group, Centre of Macromolecular Chemistry (CMaC), Department of Organic and Macromolecular Chemistry, Faculty of Sciences, Ghent University, Krijgslaan 281, Building S4, Ghent 9000, Belgium
| | - Louis Van der Meeren
- Nano-biotechnology Laboratory, Department of Biotechnology, Faculty of Bioscience Engineering, Ghent university, Proeftuinstraat 86, Ghent 9000, Belgium
| | - Anna Szabó
- Polymer Chemistry and Biomaterials (PBM) Group, Centre of Macromolecular Chemistry (CMaC), Department of Organic and Macromolecular Chemistry, Faculty of Sciences, Ghent University, Krijgslaan 281, Building S4, Ghent 9000, Belgium
| | - Andre G Skirtach
- Nano-biotechnology Laboratory, Department of Biotechnology, Faculty of Bioscience Engineering, Ghent university, Proeftuinstraat 86, Ghent 9000, Belgium
| | - Ruslan I Dmitriev
- Tissue Engineering and Biomaterials Group, Department of Human Structure and Repair, Faculty of Medical and Health Sciences, Ghent University, C. Heymanslaan 10, Ghent 9000, Belgium
| | - Janis Locs
- Institute of Biomaterials and Bioengineering, Faculty of Natural Sciences and Technology, Riga Technical University, Pulka 3, Riga LV-1007, Latvia
- Baltic Biomaterials Centre of Excellence, Headquarters at Riga Technical University, Riga LV-1658, Latvia
| | - Sandra Van Vlierberghe
- Polymer Chemistry and Biomaterials (PBM) Group, Centre of Macromolecular Chemistry (CMaC), Department of Organic and Macromolecular Chemistry, Faculty of Sciences, Ghent University, Krijgslaan 281, Building S4, Ghent 9000, Belgium
| |
Collapse
|
13
|
Huang L, Guo Z, Yang X, Zhang Y, Liang Y, Chen X, Qiu X, Chen X. Advancements in GelMA bioactive hydrogels: Strategies for infection control and bone tissue regeneration. Theranostics 2025; 15:460-493. [PMID: 39744697 PMCID: PMC11671377 DOI: 10.7150/thno.103725] [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: 09/15/2024] [Accepted: 11/04/2024] [Indexed: 01/11/2025] Open
Abstract
Infectious bone defects present a significant clinical challenge, characterized by infection, inflammation, and subsequent bone tissue destruction. Traditional treatments, including antibiotic therapy, surgical debridement, and bone grafting, often fail to address these defects effectively. However, recent advancements in biomaterials research have introduced innovative solutions for managing infectious bone defects. GelMA, a three-dimensional network of hydrophilic polymers that can absorb and retain substantial amounts of water, has attracted considerable attention in the fields of materials science and biomedical engineering. Its distinctive properties, such as biocompatibility, responsiveness to stimuli, and customisable mechanical characteristics make GelMA an exemplary scaffold material for bone tissue engineering. This review aims to thoroughly explore the current literature on antibacterial and osteogenic strategies using GelMA hydrogels for the restoration of infected bones. It discusses their fabrication methods, biocompatibility, antibacterial effectiveness, and bioactivity. We conclude by discussing the existing challenges and future research directions in this field, with the hope of inspiring further innovations in the synthesis, modification, and application of GelMA-based hydrogels for infection control and bone tissue regeneration.
Collapse
Affiliation(s)
- Lei Huang
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, China
| | - Ziyao Guo
- SCP 11A of the International Department, Guangzhou Experimental Foreign Language School, Guangzhou, China
| | - Xiaoxia Yang
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, China
| | - Yinchun Zhang
- Department of Periodontology, Shaoxing Stomatological Hospital, Shaoxing, Zhejiang, China
| | - Yiyun Liang
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, China
| | - Xiaxue Chen
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, China
| | - Xiaoling Qiu
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, China
| | - Xuan Chen
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, China
| |
Collapse
|
14
|
Liang W, Zhou C, Liu X, Xie Q, Xia L, Liu L, Bao W, Lin H, Xiong X, Zhang H, Zheng Z, Zhao J. Current status of nano-embedded growth factors and stem cells delivery to bone for targeted repair and regeneration. J Orthop Translat 2025; 50:257-273. [PMID: 39902262 PMCID: PMC11788687 DOI: 10.1016/j.jot.2024.12.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 11/01/2024] [Accepted: 12/09/2024] [Indexed: 02/05/2025] Open
Abstract
Bone-related diseases like osteoarthritis and osteoporosis impact millions globally, affecting quality of life. Osteoporosis considerably enhances the probability of bone fractures of the wrist, hip, and spine. Enhancement and acceleration of functional bone development can be achieved through the sustained delivery of growth factors (GFs) and cells in biomaterial carriers. The delivery of bioactive compounds in a targeted, spatiotemporal way that most closely resembles the natural defect repair process can be achieved by designing the carrier system with established release kinetics. Furthermore, the carrier can serve as a substrate that mimics the extracellular matrix, facilitating osteoprogenitor cell infiltration and growth for integrative tissue healing. In this report, we explore the significance of GFs within the realm of bone and cartilage tissue engineering, encompassing their encapsulation and delivery methodologies, the kinetics of release, and their amalgamation with biomaterials and stem cells (SCs) to facilitate the mending of bone fractures. Moreover, the significance of GFs in evaluating the microenvironment of bone tissue through reciprocal signaling with cells and biomaterial scaffolds is emphasized which will serve as the foundation for prospective advances in bone and cartilage tissue engineering as well as therapeutic equipment. Nanoparticles are being used in regenerative medicine to promote bone regeneration and repair by delivering osteoinductive growth factors like BMP-2, VEGF, TGF-β. These nanocarriers allow controlled release, minimizing adverse effects and ensuring growth factors are concentrated at the injury site. They are also mixed with mesenchymal stem cells (MSCs) to improve their engraftment, differentiation, and survival. This approach is a key step in developing multi-model systems that more efficiently facilitate bone regeneration. Researchers are exploring smart nanoparticles with immunomodulatory qualities to improve bonre regeneration and reduce inflammation in injury site. Despite promising preclinical results, challenges include cost management, regulatory approval, and long term safety. However, incorporating stem cell transport and growth factors in nanoparticles could revolutionize bone regeneration and offer more personalized therapies for complex bone disorders and accidents. The translational potential of this article Stem cell transport and growth factors encapsulated in nanoparticles are becoming revolutionary methods for bone regeneration and repair. By encouraging stem cells to develop into osteoblasts, osteoinductive GFs like BMP-2, VEGF, and TGF-β can be delivered under control due to nanomaterials like nanoparticles, nanofibers, and nanotubes. By ensuring sustained release, these nanocarriers lessen adverse effects and enhance therapeutic results. In order to prove their survival and development, MCSs, which are essential for bone regeneration, are mixed with nanoparticles, frequently using scaffolds that resemble the ECM of bone. Furthermore, by adjusting to the injured environment and lowering inflammation, immunomodulatory nanostructures and stimuli-responsive nanomaterials can further maximize. While there are still shotcomings to overcome, including managing expenses, negotiating regulatory processes, and guaranteeing long-term safety, this method promises to outperform traditional bone grafting by providing quicker, more individualized, and more efficient treatments. Nano-embedded growth factors and stem cell technologies have the potential to revolutionize orthopedic therapy and significantly enhance patient outcomes with further research.
Collapse
Affiliation(s)
- Wenqing Liang
- Department of Orthopedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, 316000, China
| | - Chao Zhou
- Department of Orthopedics, Zhoushan Guanghua Hospital, Zhoushan, 316000, China
| | - Xiankun Liu
- Department of Orthopedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, 316000, China
| | - Qiong Xie
- Medical Research Center, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, 316000, China
| | - Linying Xia
- Medical Research Center, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, 316000, China
| | - Lu Liu
- Medical Research Center, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, 316000, China
| | - Wenwen Bao
- Medical Research Center, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, 316000, China
| | - Hongming Lin
- Department of Orthopedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, 316000, China
| | - Xiaochun Xiong
- Department of Orthopedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, 316000, China
| | - Hao Zhang
- Department of Orthopedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, 316000, China
| | - Zeping Zheng
- Department of Orthopedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, 316000, China
| | - Jiayi Zhao
- Department of Orthopedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, 316000, China
| |
Collapse
|
15
|
Maji S, Aliabouzar M, Quesada C, Chiravuri A, Macpherson A, Pinch A, Kazyak K, Emara Z, Abeid BA, Kent RN, Midekssa FS, Zhang M, Baker BM, Franceschi RT, Fabiilli ML. Ultrasound-generated bubbles enhance osteogenic differentiation of mesenchymal stromal cells in composite collagen hydrogels. Bioact Mater 2025; 43:82-97. [PMID: 39345992 PMCID: PMC11439547 DOI: 10.1016/j.bioactmat.2024.09.018] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 08/30/2024] [Accepted: 09/13/2024] [Indexed: 10/01/2024] Open
Abstract
Hydrogels can improve the delivery of mesenchymal stromal cells (MSCs) by providing crucial biophysical cues that mimic the extracellular matrix. The differentiation of MSCs is dependent on biophysical cues like stiffness and viscoelasticity, yet conventional hydrogels cannot be dynamically altered after fabrication and implantation to actively direct differentiation. We developed a composite hydrogel, consisting of type I collagen and phase-shift emulsion, where osteogenic differentiation of MSCs can be non-invasively modulated using ultrasound. When exposed to ultrasound, the emulsion within the hydrogel was non-thermally vaporized into bubbles, which locally compacted and stiffened the collagen matrix surrounding each bubble. Bubble growth and matrix compaction were correlated, with collagen regions proximal (i.e., ≤ ∼60 μm) to the bubble displaying a 2.5-fold increase in Young's modulus compared to distal regions (i.e., > ∼60 μm). The viability and proliferation of MSCs, which were encapsulated within the composite hydrogel, were not impacted by bubble formation. In vitro and in vivo studies revealed encapsulated MSCs exhibited significantly elevated levels of RUNX2 and osteocalcin, markers of osteogenic differentiation, in collagen regions proximal to the bubble compared to distal regions. Additionally, alkaline phosphatase activity and calcium deposition were enhanced adjacent to the bubble. An opposite trend was observed for CD90, a marker of MSC stemness. Following subcutaneous implantation, bubbles persisted in the hydrogels for two weeks, which led to localized collagen alignment and increases in nuclear asymmetry. These results are a significant step toward controlling the 3D differentiation of MSCs in a non-invasive and on-demand manner.
Collapse
Affiliation(s)
- Somnath Maji
- Department of Radiology, University of Michigan, Ann Arbor, MI, USA
| | - Mitra Aliabouzar
- Department of Radiology, University of Michigan, Ann Arbor, MI, USA
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Carole Quesada
- Department of Radiology, University of Michigan, Ann Arbor, MI, USA
| | - Anjali Chiravuri
- Department of Radiology, University of Michigan, Ann Arbor, MI, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Aidan Macpherson
- Department of Radiology, University of Michigan, Ann Arbor, MI, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Abigail Pinch
- Department of Radiology, University of Michigan, Ann Arbor, MI, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Karsyn Kazyak
- Department of Radiology, University of Michigan, Ann Arbor, MI, USA
| | - Ziyad Emara
- Department of Radiology, University of Michigan, Ann Arbor, MI, USA
| | - Bachir A Abeid
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Robert N Kent
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Firaol S Midekssa
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Man Zhang
- Department of Radiology, University of Michigan, Ann Arbor, MI, USA
| | - Brendon M Baker
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Renny T Franceschi
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
- Department of Periodontics and Oral Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Mario L Fabiilli
- Department of Radiology, University of Michigan, Ann Arbor, MI, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
- Applied Physics Program, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
16
|
Yang W, Zou Q, Wang C, Ren Y, Zhang R, Lin M, Huang Z, Huangfu M, Lin L, Li W, Li X. Enhancing Bone Regeneration and Osteogenic Quality by n-HA Internalized Osteoblasts Synergized with ON Protein: Mechanistic Insights. ACS APPLIED MATERIALS & INTERFACES 2024; 16:68967-68982. [PMID: 39638777 DOI: 10.1021/acsami.4c16045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/07/2024]
Abstract
Bone scaffolds offer hope for oral jawbone repair, yet improving their osteogenic performance remains a clinical challenge. This study investigates a novel approach to enhance early bone formation and osteogenic quality by coloading hydroxyapatite (HA)─internalized osteoblasts (OHA) and osteonectin (ON) onto various scaffolds. Our findings demonstrated that the OHA could effectively facilitate the early bone regeneration by providing rapid calcium and phosphorus ion release via lysosome-mediated HA degradation, while the ON protein helps in ion deposition, cell proliferation, and matrix mineralization. When the PHA (PCL+HA) scaffold was incorporated with both the OHA and ON, the scaffold exhibited superior pro-osteogenic performance, driven by synergistic effects of rapid ion release from the OHA, slow ion release from the PHA, and upregulation of osteogenesis-related genes. The analyses of mechanisms revealed that the OHA activated MAPK and PI3K-Akt pathways, while ON stimulated calcium and Wnt signaling, collectively promoting the osteogenic potential. The strategy presented in this study paves a promising way for the development of advanced bone scaffolds to improve the bone regeneration quality.
Collapse
Affiliation(s)
- Wei Yang
- State Key Laboratory of Oral Diseases, National Center for Stomatology, and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Qin Zou
- Research Center for Nano Biomaterials, Analytical & Testing Center, Sichuan University, Chengdu, Sichuan 610064, China
| | - Chenxin Wang
- Research Center for Nano Biomaterials, Analytical & Testing Center, Sichuan University, Chengdu, Sichuan 610064, China
| | - Yuankun Ren
- State Key Laboratory of Oral Diseases, National Center for Stomatology, and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Rui Zhang
- Research Center for Nano Biomaterials, Analytical & Testing Center, Sichuan University, Chengdu, Sichuan 610064, China
| | - Mingyue Lin
- Research Center for Nano Biomaterials, Analytical & Testing Center, Sichuan University, Chengdu, Sichuan 610064, China
| | - Zeyu Huang
- State Key Laboratory of Oral Diseases, National Center for Stomatology, and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Mengxin Huangfu
- State Key Laboratory of Oral Diseases, National Center for Stomatology, and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Lili Lin
- State Key Laboratory of Oral Diseases, National Center for Stomatology, and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Wei Li
- State Key Laboratory of Oral Diseases, National Center for Stomatology, and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Xiyu Li
- State Key Laboratory of Oral Diseases, National Center for Stomatology, and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| |
Collapse
|
17
|
Sharun K, Banu SA, Alifsha B, Abualigah L, Pawde AM, Dhama K, Pal A. Mesenchymal stem cell therapy in veterinary ophthalmology: clinical evidence and prospects. Vet Res Commun 2024; 48:3517-3531. [PMID: 39212813 DOI: 10.1007/s11259-024-10522-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024]
Abstract
Mesenchymal stem cell (MSC) therapy presents a promising strategy for treating various ocular conditions in veterinary medicine. This review explores the therapeutic potential of MSCs in managing corneal ulcers, immune-mediated keratitis, chronic superficial keratitis, keratoconjunctivitis sicca, retinal degeneration, and ocular burns in feline, equine, and canine patients. Studies have demonstrated the immunomodulatory and regenerative properties of MSCs, highlighting their ability to mitigate inflammation and promote tissue regeneration. Experimental studies have shown the potential of MSC therapy in reducing corneal opacity and vascularization, indicating significant therapeutic advantages. Delivery methods play a crucial role in optimizing the therapeutic efficacy of MSCs in ocular diseases. Various delivery methods, such as intravitreal injection, subconjunctival injection, topical administration, and scaffold-mediated delivery, are being explored to optimize MSC delivery to the target ocular tissues. Clinical trials have shown significant improvements in clinical signs following MSC therapy, underscoring its efficacy in treating ocular diseases. Additionally, tissue engineering approaches incorporating MSCs, growth factors, and scaffolds offer innovative strategies for corneal regeneration and tissue repair. Despite challenges such as standardization of protocols and long-term safety assessment, ongoing research endeavours seek to unlock the full therapeutic potential of MSC therapy in ocular diseases. Future prospects in MSC therapy involve exploring scaffold and hydrogel-based approaches and cell-free therapies leveraging the bioactive molecules released by MSCs. Continued research and development efforts are essential to unlock the full therapeutic potential of MSCs and realize their transformative impact on ocular diseases in veterinary patients.
Collapse
Affiliation(s)
- Khan Sharun
- Division of Surgery, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh, India.
- Graduate Institute of Medicine, Yuan Ze University, Taoyuan, 32003, Taiwan.
| | - S Amitha Banu
- Division of Surgery, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh, India
| | - B Alifsha
- Division of Surgery, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh, India
| | - Laith Abualigah
- Computer Science Department, Al al-Bayt University, Mafraq, 25113, Jordan
- MEU Research Unit, Middle East University, Amman, 11831, Jordan
- Applied Science Research Center, Applied Science Private University, Amman, 11931, Jordan
- Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, Punjab, 140401, India
- Artificial Intelligence and Sensing Technologies (AIST) Research Center, University of Tabuk, 71491, Tabuk, Saudi Arabia
| | - A M Pawde
- Division of Surgery, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh, India
| | - Kuldeep Dhama
- Division of Pathology, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh, India
| | - Amar Pal
- Division of Surgery, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh, India
| |
Collapse
|
18
|
Chen X, Xu Z, Gao Y, Chen Y, Yin W, Liu Z, Cui W, Li Y, Sun J, Yang Y, Ma W, Zhang T, Tian T, Lin Y. Framework Nucleic Acid-Based Selective Cell Catcher for Endogenous Stem Cell Recruitment. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2406118. [PMID: 39543443 DOI: 10.1002/adma.202406118] [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: 04/29/2024] [Revised: 09/23/2024] [Indexed: 11/17/2024]
Abstract
Cell-surface engineering holds great promise in boosting endogenous stem cell attraction for tissue regeneration. However, challenges such as cellular internalization of ligand and the dynamic nature of cell membranes often complicate ligand-receptor interactions. The aim of this study is to harness the innovative potential of programmable tetrahedral framework nucleic acid (tFNA) to enable precise, tunable ligand-receptor interactions, thereby improving stem cell recruitment efficiency. This approach involves experimental screening and theoretical analysis using dissipative particle dynamics. The results demonstrate that altering the flexibility and topology of ligands on tFNA changes their cellular internalization and membrane binding efficiency. Furthermore, optimizing the distribution of the mesenchymal stem cell (MSC)-binding aptamer 19S (Apt19S) on the tFNA enhances the stem cell capture efficiency. Following successful in vitro MSC capture, Apt19S-modified tFNA is chemically linked to a hyaluronic acid hydrogel, forming an efficient "stem cell catcher" system. Subsequent in vivo experiments demonstrate that this system effectively promotes early stem cell recruitment and accelerates bone regeneration in different bone healing scenarios, including cranial and maxillary defects.
Collapse
Affiliation(s)
- Xingyu Chen
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, P. R. China
| | - Ziang Xu
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, P. R. China
| | - Yang Gao
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, P. R. China
| | - Ye Chen
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, P. R. China
| | - Wumeng Yin
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, P. R. China
| | - Zhiqiang Liu
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, P. R. China
| | - Weitong Cui
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, P. R. China
| | - Yong Li
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, P. R. China
| | - Jiafei Sun
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, P. R. China
| | - Yuting Yang
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, P. R. China
| | - Wenjuan Ma
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, P. R. China
| | - Tao Zhang
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, P. R. China
| | - Taoran Tian
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, P. R. China
| | - Yunfeng Lin
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, P. R. China
- Sichuan Provincial Engineering Research Center of Oral Biomaterials, Chengdu, Sichuan, 610041, China
| |
Collapse
|
19
|
Kim M, Wang X, Li Y, Lin Z, Collins CP, Liu Y, Ahn Y, Tsal HM, Song JW, Duan C, Zhu Y, Sun C, He TC, Luo Y, Reid RR, Ameer GA. Personalized composite scaffolds for accelerated cell- and growth factor-free craniofacial bone regeneration. Bioact Mater 2024; 41:427-439. [PMID: 39188380 PMCID: PMC11345904 DOI: 10.1016/j.bioactmat.2024.07.029] [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: 01/29/2024] [Revised: 07/16/2024] [Accepted: 07/21/2024] [Indexed: 08/28/2024] Open
Abstract
Approaches to regenerating bone often rely on integrating biomaterials and biological signals in the form of cells or cytokines. However, from a translational point of view, these approaches are challenging due to the sourcing and quality of the biologic, unpredictable immune responses, complex regulatory paths, and high costs. We describe a simple manufacturing process and a material-centric 3D-printed composite scaffold system (CSS) that offers distinct advantages for clinical translation. The CSS comprises a 3D-printed porous polydiolcitrate-hydroxyapatite composite elastomer infused with a polydiolcitrate-graphene oxide hydrogel composite. Using a micro-continuous liquid interface production 3D printer, we fabricate a precise porous ceramic scaffold with 60 wt% hydroxyapatite resembling natural bone. The resulting scaffold integrates with a thermoresponsive hydrogel composite in situ to fit the defect, which is expected to enhance surface contact with surrounding tissue and facilitate biointegration. The antioxidative properties of citrate polymers prevent long-term inflammatory responses. The CSS stimulates osteogenesis in vitro and in vivo. Within 4 weeks in a calvarial critical-sized bone defect model, the CSS accelerated ECM deposition (8-fold) and mineralized osteoid (69-fold) compared to the untreated. Through spatial transcriptomics, we demonstrated the comprehensive biological processes of CSS for prompt osseointegration. Our material-centric approach delivers impressive osteogenic properties and streamlined manufacturing advantages, potentially expediting clinical application for bone reconstruction surgeries.
Collapse
Affiliation(s)
- Mirae Kim
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, 60208, USA
- Center for Advanced Regenerative Engineering, Northwestern University, Evanston, IL, 60208, USA
| | - Xinlong Wang
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, 60208, USA
- Center for Advanced Regenerative Engineering, Northwestern University, Evanston, IL, 60208, USA
| | - Yiming Li
- Department of Preventive Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Zitong Lin
- Department of Preventive Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Caralyn P. Collins
- Center for Advanced Regenerative Engineering, Northwestern University, Evanston, IL, 60208, USA
- Department of Mechanical Engineering, Northwestern University, Evanston, IL, 60208 USA
| | - Yugang Liu
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, 60208, USA
- Center for Advanced Regenerative Engineering, Northwestern University, Evanston, IL, 60208, USA
| | - Yujin Ahn
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61820, USA
| | - Hsiu-Ming Tsal
- Department of Radiology, The University of Chicago, Chicago, IL, 60637, USA
| | - Joseph W. Song
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, 60208, USA
- Center for Advanced Regenerative Engineering, Northwestern University, Evanston, IL, 60208, USA
| | - Chongwen Duan
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, 60208, USA
- Center for Advanced Regenerative Engineering, Northwestern University, Evanston, IL, 60208, USA
| | - Yi Zhu
- Molecular Oncology Laboratory, Department of Orthopedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA
| | - Cheng Sun
- Center for Advanced Regenerative Engineering, Northwestern University, Evanston, IL, 60208, USA
- Department of Mechanical Engineering, Northwestern University, Evanston, IL, 60208 USA
| | - Tong-Chuan He
- Center for Advanced Regenerative Engineering, Northwestern University, Evanston, IL, 60208, USA
- Molecular Oncology Laboratory, Department of Orthopedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA
| | - Yuan Luo
- Department of Preventive Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
- Clinical and Translational Sciences Institute, Northwestern University, Chicago, IL, 60611, USA
- Center for Collaborative AI in Healthcare, Institute for AI in Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Russell R. Reid
- Center for Advanced Regenerative Engineering, Northwestern University, Evanston, IL, 60208, USA
- Laboratory of Craniofacial Biology and Development, Section of Plastic and Reconstructive Surgery, Department of Surgery, The University of Chicago Medical Center, Chicago, IL, 60637, USA
| | - Guillermo A. Ameer
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, 60208, USA
- Center for Advanced Regenerative Engineering, Northwestern University, Evanston, IL, 60208, USA
- Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
- Chemistry of Life Process Institute, Northwestern University, Chicago, IL, 60208, USA
- International Institute for Nanotechnology, Northwestern University, Evanston, IL, 60208, USA
| |
Collapse
|
20
|
Zhang X, Yang B, Feng L, Xu X, Wang C, Lee YW, Wang M, Lu X, Qin L, Lin S, Bian L, Li G. Augmenting osteoporotic bone regeneration through a hydrogel-based rejuvenating microenvironment. Bioact Mater 2024; 41:440-454. [PMID: 39188381 PMCID: PMC11347042 DOI: 10.1016/j.bioactmat.2024.07.036] [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: 04/30/2024] [Revised: 07/29/2024] [Accepted: 07/30/2024] [Indexed: 08/28/2024] Open
Abstract
Osteoporotic bone defects pose a significant challenge for bone regeneration as they exhibit impaired healing capacity and delayed healing period. To address this issue, this study introduces a hydrogel that creates a rejuvenating microenvironment, thereby facilitating efficient bone repair during the initial two weeks following bone defect surgery. The hydrogel, named GelHFS, was created through host-guest polymerization of gelatin and acrylated β-cyclodextrin. Incorporation of the human fetal mesenchymal stem cell secretome (HFS) formed GelHFS hydrogel aimed at mimicking a rejuvenated stem cell niche. Our results demonstrated that GelHFS hydrogel promotes cell stellate spreading and osteogenic differentiation via integrin β1-induced focal adhesion pathway. Implantation of GelHFS hydrogel in an osteoporotic bone defect rat model recruited endogenous integrin β1-expressing cells and enhanced new bone formation and bone strength. Our findings reveal that GelHFS hydrogel provides a rejuvenating niche for endogenous MSCs and enhances bone regeneration in osteoporotic bone defect. These findings highlight the potential of GelHFS hydrogel as an effective therapeutic strategy for addressing challenging bone healing such as osteoporotic bone regeneration.
Collapse
Affiliation(s)
- Xiaoting Zhang
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
- Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
- The CUHK-ACC Space Medicine Centre on Health Maintenance of Musculoskeletal System, The Chinese University of Hong Kong Shenzhen Research Institute, Shenzhen, PR China
| | - Boguang Yang
- Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
| | - Lu Feng
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
- Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
- The CUHK-ACC Space Medicine Centre on Health Maintenance of Musculoskeletal System, The Chinese University of Hong Kong Shenzhen Research Institute, Shenzhen, PR China
| | - Xiayi Xu
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 511442, PR China
| | - Chenmin Wang
- Department of Orthopaedics and Traumatology, The University of Hong Kong, Hong Kong, China
| | - Yuk-wai Lee
- SH Ho Scoliosis Research Laboratory, Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong, China
- Joint Scoliosis Research Center of the Chinese University of Hong Kong and Nanjing University, The Chinese University of Hong Kong, Hong Kong, China
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, China
| | - Ming Wang
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
- Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
- The CUHK-ACC Space Medicine Centre on Health Maintenance of Musculoskeletal System, The Chinese University of Hong Kong Shenzhen Research Institute, Shenzhen, PR China
| | - Xuan Lu
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
- Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
- The CUHK-ACC Space Medicine Centre on Health Maintenance of Musculoskeletal System, The Chinese University of Hong Kong Shenzhen Research Institute, Shenzhen, PR China
| | - Ling Qin
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
- Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
| | - Sien Lin
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
- Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
- The CUHK-ACC Space Medicine Centre on Health Maintenance of Musculoskeletal System, The Chinese University of Hong Kong Shenzhen Research Institute, Shenzhen, PR China
| | - Liming Bian
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 511442, PR China
| | - Gang Li
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
- Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
- The CUHK-ACC Space Medicine Centre on Health Maintenance of Musculoskeletal System, The Chinese University of Hong Kong Shenzhen Research Institute, Shenzhen, PR China
| |
Collapse
|
21
|
Hu X, Yang S, Zhao W, Zhang Z, Qiao L, Wu H, Su Q, Che L, Zhou K, Li K, He J. Novel multi-functional microsphere scaffold with shape memory function for bone regeneration. BIOMATERIALS ADVANCES 2024; 163:213958. [PMID: 39053385 DOI: 10.1016/j.bioadv.2024.213958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 07/09/2024] [Accepted: 07/12/2024] [Indexed: 07/27/2024]
Abstract
Irregular bone defects caused by trauma and bone diseases provide a complex implant environment for surgery. Traditional implants often fail to integrate well with the surrounding bone defect interface, therefore, developing an artificial bone scaffold that can adapt to irregular bone defect boundaries is of significant importance for bone defect repair. This study successfully utilized a shape memory ternary copolymer polylactic acid-trimethylene carbonate-hydroxyacetic acid (PLLA-TMC-GA) and dopamine-modified nano-hydroxyapatite (PHA) composite to construct a temperature-responsive bone repair scaffold (PTG/PHA), thereby enhancing the interface compatibility between the implant and the surrounding environment. The addition of PHA has effectively improved the hydrophilicity of the stent and significantly increased its mechanical strength. Furthermore, the Sodium alginate (SA) hydrogel loaded with Icariin (Ica) coated on the stent surface promotes the growth and differentiation of bone cells through the drug-scaffold synergistic effect. Both in vivo and in vitro experiments have shown that the synergistic effect of the composite stent with Icariin significantly enhances the repair of bone defects. This study provides a promising tissue engineering method for the repair of irregular bone defects.
Collapse
Affiliation(s)
- Xulin Hu
- Clinical Medical College and Affiliated Hospital of Chengdu University, Chengdu University, Chengdu, Sichuan 610081, China; Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Shuhao Yang
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400042, China
| | - Weiming Zhao
- Clinical Medical College and Affiliated Hospital of Chengdu University, Chengdu University, Chengdu, Sichuan 610081, China
| | - Zhen Zhang
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Liang Qiao
- Henan Univ Sci & Technol, Affiliated Hosp 1, Key Lab Neuromol Biol, Coll Clin Med, Luoyang, Henan 471003, China
| | - Haoming Wu
- Clinical Medical College and Affiliated Hospital of Chengdu University, Chengdu University, Chengdu, Sichuan 610081, China
| | - Qiao Su
- West China School of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Lanyu Che
- Clinical Medical College and Affiliated Hospital of Chengdu University, Chengdu University, Chengdu, Sichuan 610081, China
| | - Kai Zhou
- Department of Orthopedics and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China.
| | - Kainan Li
- Clinical Medical College and Affiliated Hospital of Chengdu University, Chengdu University, Chengdu, Sichuan 610081, China.
| | - Jian He
- College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, Henan 471000, China.
| |
Collapse
|
22
|
Sun L, Chen J, Li LJ, Li L. Similarity-based metric analysis approach for predicting osteogenic differentiation correlation coefficients and discovering the novel osteogenic-related gene FOXA1 in BMSCs. PeerJ 2024; 12:e18068. [PMID: 39308804 PMCID: PMC11416762 DOI: 10.7717/peerj.18068] [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: 06/04/2024] [Accepted: 08/19/2024] [Indexed: 09/25/2024] Open
Abstract
Background As a powerful tool, bioinformatics analysis is playing an increasingly important role in many fields. Osteogenic differentiation is a complex biological process involving the fine regulation of numerous genes and signaling pathways. Method Osteogenic differentiation-related genes are collected from the online databases. Then, we proposed two indexes Jaccard similarity and Sorensen-Dice similarity to measure the topological relevance of genes in the human PPI network. Furthermore, we selected three pathways involving osteoblast-related transcription factors, osteoblast differentiation, and RUNX2 regulation of osteoblast differentiation for investigation. Subsequently, we performed functional a enrichment analysis of these top-ranked genes to check whether these candidate genes identified by similarity-based metrics are enriched in some specific biological functions and states. we performed a permutation test to investigate the similarity score with four well-known osteogenic differentiation-related pathways including hedgehog signaling pathway, BMP signaling, ERK pathway, and Wnt signaling pathway to check whether these osteogenic differentiation-related pathways can be regulated by FOXA1. Lentiviral transfection was used to knockdown and overexpress gene FOXA1 in human bone mesenchymal stem cells (hBMSCs). Alkaline phosphatase (ALP) staining and Alizarin Red staining (ARS) were employed to investigate osteogenic differentiation of hBMSCs. Result After data collection, human PPI network involving 19,344 genes is included in our analysis. After simplifying, we used Jaccard and Sorensen-Dice similarity to identify osteogenic differentiation-related genes and integrated into a final similarity matrix. Furthermore, we calculated the sum of similarity scores with these osteogenic differentiation-related genes for each gene and found 337 osteogenic differentiation-related genes are involved in our analysis. We selected three pathways involving osteoblast-related transcription factors, osteoblast differentiation, and RUNX2 regulation of osteoblast differentiation for investigation and performed functional enrichment analysis of these top-ranked 50 genes. The results collectively demonstrate that these candidate genes can indeed capture osteogenic differentiation-related features of hBSMCs. According to the novel analyzing method, we found that these four pathways have significantly higher similarity with FOXA1 than random noise. Moreover, knockdown FOXA1 significantly increased the ALP activity and mineral deposits. Furthermore, overexpression of FOXA1 dramatically decreased the ALP activity and mineral deposits. Conclusion In summary, this study showed that FOXA1 is a novel significant osteogenic differentiation-related transcription factor. Moreover, our study has tightly integrated bioinformatics analysis with biological knowledge, and developed a novel method for analyzing the osteogenic differentiation regulatory network.
Collapse
Affiliation(s)
- Lingtong Sun
- Hangzhou Xixi Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Juan Chen
- Hangzhou Xixi Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Li Jun Li
- Hangzhou Xixi Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Lingdi Li
- Department of Medical Oncology, Hangzhou Cancer Hospital, Hangzhou, Zhejiang, China
| |
Collapse
|
23
|
Zhao L, Wang L, Huang J, Chen H, Liu L, Shi M, Zhang M. Label-Free Imaging of Mesenchymal Stem Cell Spheroid Differentiation with Flexible-Probe SECM and a Microfluidic Device. Anal Chem 2024; 96:13473-13481. [PMID: 39122667 DOI: 10.1021/acs.analchem.4c01637] [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: 08/12/2024]
Abstract
Mesenchymal stem cells (MSCs) have emerged as an indispensable source for stem cell research and preclinical studies due to their capacity for in vitro proliferation and their potential to differentiate into mesodermal lineages, particularly into osteoblasts. This capability has propelled their application in the fields of bone regeneration and osteochondral repair. Traditional methodologies for assessing the differentiation status of MSCs necessitate invasive procedures such as cell lysis or fixation. In this study, we introduce a nondestructive technique that utilizes an integrated label-free approach to evaluate the osteogenic maturation of MSC spheroid aggregates. This method employs scanning electrochemical microscopy (SECM) with a flexible probe in conjunction with a top-removable microfluidic device designed for easy SECM access. By tracking the production rate of p-aminophenol (PAP) in the generation/collection mode and assessing morphological changes via the negative feedback mode using [Ru(NH3)6]Cl3 (Ruhex), we can discern variations in the alkaline phosphatase (ALP) activity indicative of osteogenic differentiation. This innovative strategy enables the direct evaluation of osteogenic differentiation in MSC spheroids cultured within microwell arrays without necessitating any labeling procedures. The utilization of a flexible microelectrode as the probe that scans in contact mode (with probe-substrate distances potentially as minimal as 0 μm) affords enhanced resolution compared to the traditional stiff-probe technique. Furthermore, this method is compatible with subsequent molecular biology assays, including gene expression analysis and immunofluorescence, thereby confirming the electrochemical findings and establishing the validity of this integrative approach.
Collapse
Affiliation(s)
- Liang Zhao
- Center of Excellence for Environmental Safety and Biological Effects, College of Chemistry and Life Science, Department of Chemistry, Beijing University of Technology, 100 Pingleyuan, Beijing 100124, China
- Beijing Key Laboratory for Bioengineering and Sensing Technology, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, 30 Xueyuan Road, Beijing 100083, China
| | - Lin Wang
- Beijing Key Laboratory for Bioengineering and Sensing Technology, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, 30 Xueyuan Road, Beijing 100083, China
| | - Jing Huang
- Beijing Key Laboratory for Bioengineering and Sensing Technology, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, 30 Xueyuan Road, Beijing 100083, China
| | - Hongyu Chen
- Beijing Key Laboratory for Bioengineering and Sensing Technology, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, 30 Xueyuan Road, Beijing 100083, China
| | - Lu Liu
- Beijing Key Laboratory for Bioengineering and Sensing Technology, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, 30 Xueyuan Road, Beijing 100083, China
| | - Mi Shi
- Beijing Key Laboratory for Bioengineering and Sensing Technology, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, 30 Xueyuan Road, Beijing 100083, China
| | - Meiqin Zhang
- Beijing Key Laboratory for Bioengineering and Sensing Technology, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, 30 Xueyuan Road, Beijing 100083, China
| |
Collapse
|
24
|
Yang K, Wu Z, Zhang K, Weir MD, Xu HHK, Cheng L, Huang X, Zhou W. Unlocking the potential of stimuli-responsive biomaterials for bone regeneration. Front Pharmacol 2024; 15:1437457. [PMID: 39144636 PMCID: PMC11322102 DOI: 10.3389/fphar.2024.1437457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 07/18/2024] [Indexed: 08/16/2024] Open
Abstract
Bone defects caused by tumors, osteoarthritis, and osteoporosis attract great attention. Because of outstanding biocompatibility, osteogenesis promotion, and less secondary infection incidence ratio, stimuli-responsive biomaterials are increasingly used to manage this issue. These biomaterials respond to certain stimuli, changing their mechanical properties, shape, or drug release rate accordingly. Thereafter, the activated materials exert instructive or triggering effects on cells and tissues, match the properties of the original bone tissues, establish tight connection with ambient hard tissue, and provide suitable mechanical strength. In this review, basic definitions of different categories of stimuli-responsive biomaterials are presented. Moreover, possible mechanisms, advanced studies, and pros and cons of each classification are discussed and analyzed. This review aims to provide an outlook on the future developments in stimuli-responsive biomaterials.
Collapse
Affiliation(s)
- Ke Yang
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Lab of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
- Clinical Research Center for Oral Tissue Deficiency Diseases of Fujian Province, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Zhuoshu Wu
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Lab of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
- Clinical Research Center for Oral Tissue Deficiency Diseases of Fujian Province, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Keke Zhang
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China
| | - Michael D. Weir
- Department of Biomaterials and Regenerative Dental Medicine, University of Maryland School of Dentistry, Baltimore, MD, United States
| | - Hockin H. K. Xu
- Department of Biomaterials and Regenerative Dental Medicine, University of Maryland School of Dentistry, Baltimore, MD, United States
| | - Lei Cheng
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China School of Stomatology & Department of Operative Dentistry and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xiaojing Huang
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Lab of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
- Clinical Research Center for Oral Tissue Deficiency Diseases of Fujian Province, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Wen Zhou
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Lab of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
- Clinical Research Center for Oral Tissue Deficiency Diseases of Fujian Province, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| |
Collapse
|
25
|
Mahajan A, Sharma S, Bhadada SK, Aggarwal A, Bhattacharyya S. Engineering a 3-dimensional tissue construct with adipose-derived stem cells for healing bone defect: An ex vivo study with femur head. Biotechnol J 2024; 19:e2300751. [PMID: 38987220 DOI: 10.1002/biot.202300751] [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: 12/30/2023] [Revised: 05/23/2024] [Accepted: 06/12/2024] [Indexed: 07/12/2024]
Abstract
The compatibility of bone graft substitutes (BGS) with mesenchymal stem cells (MSCs) is an important parameter to consider for their use in repairing bone defects as it eventually affects the clinical outcome. In the present study, a few commercially available BGS - β-tricalcium phosphate (β-TCP), calcium sulfate, gelatin sponge, and different forms of hydroxyapatite (HAP) were screened for their interactions with MSCs from adipose tissue (ADSCs). It was demonstrated that HAP block favorably supported ADSC viability, morphology, migration, and differentiation compared to other scaffolds. The results strongly suggest the importance of preclinical evaluation of bone scaffolds for their cellular compatibility. Furthermore, the bone regenerative potential of HAP block with ADSCs was evaluated in an ex vivo bone defect model developed using patient derived trabecular bone explants. The explants were cultured for 45 days in vitro and bone formation was assessed by expression of osteogenic genes, ALP secretion, and high resolution computed tomography. Our findings confirmed active bone repair process in ex vivo settings. Addition of ADSCs significantly accelerated the repair process and improved bone microarchitecture. This ex vivo bone defect model can emerge as a viable alternative to animal experimentation and also as a potent tool to evaluate patient specific bone therapeutics under controlled conditions.
Collapse
Affiliation(s)
- Aditi Mahajan
- Department of Biophysics, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Siddhartha Sharma
- Department of Orthopedics, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Sanjay Kumar Bhadada
- Department of Endocrinology, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Aditya Aggarwal
- Department of Orthopedics, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Shalmoli Bhattacharyya
- Department of Biophysics, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| |
Collapse
|
26
|
Zhu F, Wang T, Wang G, Yan C, He B, Qiao B. The Exosome-Mediated Bone Regeneration: An Advanced Horizon Toward the Isolation, Engineering, Carrying Modalities, and Mechanisms. Adv Healthc Mater 2024; 13:e2400293. [PMID: 38426417 DOI: 10.1002/adhm.202400293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Indexed: 03/02/2024]
Abstract
Exosomes, nanoparticles secreted by various cells, composed of a bilayer lipid membrane, and containing bioactive substances such as proteins, nucleic acids, metabolites, etc., have been intensively investigated in tissue engineering owing to their high biocompatibility and versatile biofunction. However, there is still a lack of a high-quality review on bone defect regeneration potentiated by exosomes. In this review, the biogenesis and isolation methods of exosomes are first introduced. More importantly, the engineered exosomes of the current state of knowledge are discussed intensively in this review. Afterward, the biomaterial carriers of exosomes and the mechanisms of bone repair elucidated by compelling evidence are presented. Thus, future perspectives and concerns are revealed to help devise advanced modalities based on exosomes to overcome the challenges of bone regeneration. It is totally believed this review will attract special attention from clinicians and provide promising ideas for their future works.
Collapse
Affiliation(s)
- Fukang Zhu
- Department of Orthopaedics, The First Affiliated Hospital of Chongqing Medical University, 1 Youyi Rd, Chongqing, 400010, P. R. China
- Orthopedic Laboratory of Chongqing Medical University, Chongqing, 400010, P. R. China
| | - Taiyou Wang
- Department of Orthopaedics, The First Affiliated Hospital of Chongqing Medical University, 1 Youyi Rd, Chongqing, 400010, P. R. China
- Orthopedic Laboratory of Chongqing Medical University, Chongqing, 400010, P. R. China
| | - Guangjian Wang
- Department of Orthopaedics, The First Affiliated Hospital of Chongqing Medical University, 1 Youyi Rd, Chongqing, 400010, P. R. China
- Department of Orthopaedics, The People's Hospital of Rongchang District, Chongqing, 402460, P. R. China
| | - Caiping Yan
- Department of Orthopaedics, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, 401120, P. R. China
| | - Bin He
- Department of Orthopaedics, The First Affiliated Hospital of Chongqing Medical University, 1 Youyi Rd, Chongqing, 400010, P. R. China
- Orthopedic Laboratory of Chongqing Medical University, Chongqing, 400010, P. R. China
| | - Bo Qiao
- Department of Orthopaedics, The First Affiliated Hospital of Chongqing Medical University, 1 Youyi Rd, Chongqing, 400010, P. R. China
- Orthopedic Laboratory of Chongqing Medical University, Chongqing, 400010, P. R. China
| |
Collapse
|
27
|
Abdal Dayem A, Yan E, Do M, Kim Y, Lee Y, Cho SG, Kim DH. Engineering extracellular vesicles for ROS scavenging and tissue regeneration. NANO CONVERGENCE 2024; 11:24. [PMID: 38922501 PMCID: PMC11208369 DOI: 10.1186/s40580-024-00430-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 05/22/2024] [Indexed: 06/27/2024]
Abstract
Stem cell therapy holds promise for tissue regeneration, yet significant challenges persist. Emerging as a safer and potentially more effective alternative, extracellular vesicles (EVs) derived from stem cells exhibit remarkable abilities to activate critical signaling cascades, thereby facilitating tissue repair. EVs, nano-scale membrane vesicles, mediate intercellular communication by encapsulating a diverse cargo of proteins, lipids, and nucleic acids. Their therapeutic potential lies in delivering cargos, activating signaling pathways, and efficiently mitigating oxidative stress-an essential aspect of overcoming limitations in stem cell-based tissue repair. This review focuses on engineering and applying EVs in tissue regeneration, emphasizing their role in regulating reactive oxygen species (ROS) pathways. Additionally, we explore strategies to enhance EV therapeutic activity, including functionalization and incorporation of antioxidant defense proteins. Understanding these molecular mechanisms is crucial for optimizing EV-based regenerative therapies. Insights into EV and ROS signaling modulation pave the way for targeted and efficient regenerative therapies harnessing the potential of EVs.
Collapse
Affiliation(s)
- Ahmed Abdal Dayem
- Department of Stem Cell and Regenerative Biotechnology, Molecular & Cellular Reprogramming Center, Institute of Advanced Regenerative Science, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul, 05029, Republic of Korea
| | - Ellie Yan
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Minjae Do
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Yoojung Kim
- Department of Stem Cell and Regenerative Biotechnology, Molecular & Cellular Reprogramming Center, Institute of Advanced Regenerative Science, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul, 05029, Republic of Korea
| | - Yeongseo Lee
- Department of Stem Cell and Regenerative Biotechnology, Molecular & Cellular Reprogramming Center, Institute of Advanced Regenerative Science, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul, 05029, Republic of Korea
| | - Ssang-Goo Cho
- Department of Stem Cell and Regenerative Biotechnology, Molecular & Cellular Reprogramming Center, Institute of Advanced Regenerative Science, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul, 05029, Republic of Korea.
- R&D Team, StemExOne Co., Ltd., 307 KU Technology Innovation Bldg, 120, Neungdong-ro, Gwangjin- gu, Seoul, 05029, Republic of Korea.
| | - Deok-Ho Kim
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21205, USA.
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD, 21205, USA.
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, 21205, USA.
- Center for Microphysiological Systems, Johns Hopkins University, Baltimore, MD, 21205, USA.
- Institute for NanoBiotechnology, Johns Hopkins University, Baltimore, MD, 21218, USA.
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
| |
Collapse
|
28
|
Edwards SD, Ganash M, Guan Z, Lee J, Kim YJ, Jeong KJ. Enhanced osteogenesis of mesenchymal stem cells encapsulated in injectable microporous hydrogel. Sci Rep 2024; 14:14665. [PMID: 38918510 PMCID: PMC11199573 DOI: 10.1038/s41598-024-65731-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Accepted: 06/24/2024] [Indexed: 06/27/2024] Open
Abstract
Delivery of therapeutic stem cells to treat bone tissue damage is a promising strategy that faces many hurdles to clinical translation. Among them is the design of a delivery vehicle which promotes desired cell behavior for new bone formation. In this work, we describe the use of an injectable microporous hydrogel, made of crosslinked gelatin microgels, for the encapsulation and delivery of human mesenchymal stem cells (MSCs) and compared it to a traditional nonporous injectable hydrogel. MSCs encapsulated in the microporous hydrogel showed rapid cell spreading with direct cell-cell connections whereas the MSCs in the nonporous hydrogel were entrapped by the surrounding polymer mesh and isolated from each other. On a per-cell basis, encapsulation in microporous hydrogel induced a 4 × increase in alkaline phosphatase (ALP) activity and calcium mineral deposition in comparison to nonporous hydrogel, as measured by ALP and calcium assays, which indicates more robust osteogenic differentiation. RNA-seq confirmed the upregulation of the genes and pathways that are associated with cell spreading and cell-cell connections, as well as the osteogenesis in the microporous hydrogel. These results demonstrate that microgel-based injectable hydrogels can be useful tools for therapeutic cell delivery for bone tissue repair.
Collapse
Affiliation(s)
- Seth D Edwards
- Department of Chemical Engineering and Bioengineering, University of New Hampshire, Durham, NH, 03824, USA
| | - Mrinal Ganash
- Department of Chemical Engineering and Bioengineering, University of New Hampshire, Durham, NH, 03824, USA
| | - Ziqiang Guan
- Department of Chemical Engineering and Bioengineering, University of New Hampshire, Durham, NH, 03824, USA
| | - Jeil Lee
- Department of Biological and Chemical Engineering, Hongik University, Sejong City, Republic of Korea
| | - Young Jo Kim
- Department of Chemical Engineering and Bioengineering, University of New Hampshire, Durham, NH, 03824, USA
| | - Kyung Jae Jeong
- Department of Chemical Engineering and Bioengineering, University of New Hampshire, Durham, NH, 03824, USA.
| |
Collapse
|
29
|
Yang X, Zhang S, Lu J, Chen X, Zheng T, He R, Ye C, Xu J. Therapeutic potential of mesenchymal stem cell-derived exosomes in skeletal diseases. Front Mol Biosci 2024; 11:1268019. [PMID: 38903180 PMCID: PMC11187108 DOI: 10.3389/fmolb.2024.1268019] [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: 07/27/2023] [Accepted: 05/16/2024] [Indexed: 06/22/2024] Open
Abstract
Skeletal diseases impose a considerable burden on society. The clinical and tissue-engineering therapies applied to alleviate such diseases frequently result in complications and are inadequately effective. Research has shifted from conventional therapies based on mesenchymal stem cells (MSCs) to exosomes derived from MSCs. Exosomes are natural nanocarriers of endogenous DNA, RNA, proteins, and lipids and have a low immune clearance rate and good barrier penetration and allow targeted delivery of therapeutics. MSC-derived exosomes (MSC-exosomes) have the characteristics of both MSCs and exosomes, and so they can have both immunosuppressive and tissue-regenerative effects. Despite advances in our knowledge of MSC-exosomes, their regulatory mechanisms and functionalities are unclear. Here we review the therapeutic potential of MSC-exosomes for skeletal diseases.
Collapse
Affiliation(s)
- Xiaobo Yang
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, China
| | - Shaodian Zhang
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, China
| | - Jinwei Lu
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, China
| | - Xiaoling Chen
- Department of Plastic Surgery, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang, China
| | - Tian Zheng
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, China
| | - Rongxin He
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, China
| | - Chenyi Ye
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, China
| | - Jianbin Xu
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, China
| |
Collapse
|
30
|
Einabadi M, Izadyari Aghmiuni A, Foroutani L, Ai A, Namini MS, Farzin A, Nahanmoghadam A, Shirian S, Kargar Jahromi H, Ai J. Evaluation of the effect of co-transplantation of collagen-hydroxyapatite bio-scaffold containing nanolycopene and human endometrial mesenchymal stem cell derived exosomes to regenerate bone in rat critical size calvarial defect. Regen Ther 2024; 26:387-400. [PMID: 39045576 PMCID: PMC11263782 DOI: 10.1016/j.reth.2024.02.006] [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: 11/19/2023] [Revised: 02/16/2024] [Accepted: 02/25/2024] [Indexed: 07/25/2024] Open
Abstract
This study aimed to evaluate the effect of nanoparticles based on the PLGA and biomolecule of lycopene (i.e. NLcp) and exosomes loaded on hydroxyapatite/collagen-based scaffolds (HA/Coll), on human endometrial MSCs (hEnMSCs) differentiation into osteoblast cells. To this end, after synthesizing NLcp and isolating hEnMSC-derived exosomes, and studying their characterizations, HA/Coll scaffold with/without NLcp and exosome was fabricated. In following, the rat skull-defect model was created on 54 male Sprague-Dawley rats (12 weeks old) which were classified into 6 groups [control group (4 healthy rats), negative control group: bone defect without grafting (10 rats), and experimental groups including bone defect grafted with HA/Coll scaffold (10 rats), HA/Coll/NLcp scaffold (10 rats), HA/Coll scaffold + exosome (10 rats), and HA/Coll-NLcp scaffold + exosome (10 rats)]. Finally, the grafted membrane along with its surrounding tissues was removed at 90 days after surgery, to assess the amount of defect repair by Hematoxylin and eosin staining. Moreover, immunohistochemical and X-ray Micro-Computed Tomography (Micro-CT) analyses were performed to assess osteocalcin and mean bone volume fraction (BVF). Based on the results, although, the existence of the exosome in the scaffold network can significantly increase mean BVF compared to HA/Coll scaffold and HA/Coll-NLcp scaffold (2.25-fold and 1.5-fold, respectively). However, the combination of NLcp and exosome indicated more effect on mean BVF; so that the HA/Coll-NLcp scaffold + exosome led to a 15.95 % increase in mean BVF than the HA/Coll scaffold + exosome. Hence, synthesized NLcp in this study can act as a suitable bioactive to stimulate the osteogenic, promotion of cell proliferation and its differentiation when used in the polymer scaffold structure or loaded into polymeric carriers containing the exosome.
Collapse
Affiliation(s)
- Masoumeh Einabadi
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Laleh Foroutani
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Arman Ai
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mojdeh Salehi Namini
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Ali Farzin
- Department of Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Amir Nahanmoghadam
- Department of Chemical Engineering, Faculty of Engineering, Shiraz Branch, Islamic Azad University, Shiraz, Iran
| | - Sadegh Shirian
- Department of Pathology, School of Veterinary Pathology, Shahrekord University, Shahrekord, Iran
| | - Hossein Kargar Jahromi
- Research Center for Noncommunicable Diseases, Jahrom University of Medical Sciences, Jahrom, Iran
| | - Jafar Ai
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
31
|
Chen YQ, Wu MC, Wei MT, Kuo JC, Yu HW, Chiou A. High-viscosity driven modulation of biomechanical properties of human mesenchymal stem cells promotes osteogenic lineage. Mater Today Bio 2024; 26:101058. [PMID: 38681057 PMCID: PMC11046220 DOI: 10.1016/j.mtbio.2024.101058] [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: 01/15/2024] [Revised: 04/10/2024] [Accepted: 04/13/2024] [Indexed: 05/01/2024] Open
Abstract
Biomechanical cues could effectively govern cell gene expression to direct the differentiation of specific stem cell lineage. Recently, the medium viscosity has emerged as a significant mechanical stimulator that regulates the cellular mechanical properties and various physiological functions. However, whether the medium viscosity can regulate the mechanical properties of human mesenchymal stem cells (hMSCs) to effectively trigger osteogenic differentiation remains uncertain. The mechanism by which cells sense and respond to changes in medium viscosity, and regulate cell mechanical properties to promote osteogenic lineage, remains elusive. In this study, we demonstrated that hMSCs, cultured in a high-viscosity medium, exhibited larger cell spreading area and higher intracellular tension, correlated with elevated formation of actin stress fibers and focal adhesion maturation. Furthermore, these changes observed in hMSCs were associated with activation of TRPV4 (transient receptor potential vanilloid sub-type 4) channels on the cell membrane. This feedback loop among TRPV4 activation, cell spreading and intracellular tension results in calcium influx, which subsequently promotes the nuclear localization of NFATc1 (nuclear factor of activated T cells 1). Concomitantly, the elevated intracellular tension induced nuclear deformation and promoted the nuclear localization of YAP (YES-associated protein). The concurrent activation of NFATc1 and YAP significantly enhanced alkaline phosphatase (ALP) for pre-osteogenic activity. Taken together, these findings provide a more comprehensive view of how viscosity-induced alterations in biomechanical properties of MSCs impact the expression of osteogenesis-related genes, and ultimately promote osteogenic lineage.
Collapse
Affiliation(s)
- Yin-Quan Chen
- Cancer Progression Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Ming-Chung Wu
- Institute of Biochemistry and Molecular Biology, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Ming-Tzo Wei
- Department of Chemical and Biological Engineering, Princeton University, Princeton, NJ, USA
| | - Jean-Cheng Kuo
- Institute of Biochemistry and Molecular Biology, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Helen Wenshin Yu
- Institute of Biochemistry and Molecular Biology, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Arthur Chiou
- Institute of Biophotonics, National Yang Ming Chiao Tung University, Taipei, Taiwan
| |
Collapse
|
32
|
Wu S, Gai T, Chen J, Chen X, Chen W. Smart responsive in situ hydrogel systems applied in bone tissue engineering. Front Bioeng Biotechnol 2024; 12:1389733. [PMID: 38863497 PMCID: PMC11165218 DOI: 10.3389/fbioe.2024.1389733] [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: 02/22/2024] [Accepted: 04/15/2024] [Indexed: 06/13/2024] Open
Abstract
The repair of irregular bone tissue suffers severe clinical problems due to the scarcity of an appropriate therapeutic carrier that can match dynamic and complex bone damage. Fortunately, stimuli-responsive in situ hydrogel systems that are triggered by a special microenvironment could be an ideal method of regenerating bone tissue because of the injectability, in situ gelatin, and spatiotemporally tunable drug release. Herein, we introduce the two main stimulus-response approaches, exogenous and endogenous, to forming in situ hydrogels in bone tissue engineering. First, we summarize specific and distinct responses to an extensive range of external stimuli (e.g., ultraviolet, near-infrared, ultrasound, etc.) to form in situ hydrogels created from biocompatible materials modified by various functional groups or hybrid functional nanoparticles. Furthermore, "smart" hydrogels, which respond to endogenous physiological or environmental stimuli (e.g., temperature, pH, enzyme, etc.), can achieve in situ gelation by one injection in vivo without additional intervention. Moreover, the mild chemistry response-mediated in situ hydrogel systems also offer fascinating prospects in bone tissue engineering, such as a Diels-Alder, Michael addition, thiol-Michael addition, and Schiff reactions, etc. The recent developments and challenges of various smart in situ hydrogels and their application to drug administration and bone tissue engineering are discussed in this review. It is anticipated that advanced strategies and innovative ideas of in situ hydrogels will be exploited in the clinical field and increase the quality of life for patients with bone damage.
Collapse
Affiliation(s)
- Shunli Wu
- College of Marine Life Sciences, Ocean University of China, Qingdao, China
- Hangzhou Singclean Medical Products Co., Ltd, Hangzhou, China
| | - Tingting Gai
- School of Medicine, Shanghai University, Shanghai, China
| | - Jie Chen
- Jiaxing Vocational Technical College, Department of Student Affairs, Jiaxing, China
| | - Xiguang Chen
- College of Marine Life Science, Ocean University of China, Qingdao, China
- Laoshan Laboratory, Qingdao, China
| | - Weikai Chen
- Department of Orthopedics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
33
|
Rodham P, Khaliq F, Giannoudis V, Giannoudis PV. Cellular therapies for bone repair: current insights. J Orthop Traumatol 2024; 25:28. [PMID: 38789881 PMCID: PMC11132192 DOI: 10.1186/s10195-024-00768-0] [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: 12/04/2023] [Accepted: 05/01/2024] [Indexed: 05/26/2024] Open
Abstract
Mesenchymal stem cells are core to bone homeostasis and repair. They both provide the progenitor cells from which bone cells are formed and regulate the local cytokine environment to create a pro-osteogenic environment. Dysregulation of these cells is often seen in orthopaedic pathology and can be manipulated by the physician treating the patient. This narrative review aims to describe the common applications of cell therapies to bone healing whilst also suggesting the future direction of these techniques.
Collapse
Affiliation(s)
- Paul Rodham
- Academic Department of Trauma and Orthopaedics, School of Medicine, University of Leeds, Leeds, UK
| | - Farihah Khaliq
- Academic Department of Trauma and Orthopaedic Surgery, School of Medicine, University of Leeds, Leeds, UK
| | - Vasileos Giannoudis
- Academic Department of Trauma and Orthopaedics, School of Medicine, University of Leeds, Leeds, UK
| | - Peter V Giannoudis
- Academic Department of Trauma and Orthopaedics, School of Medicine, University of Leeds, Leeds, UK.
- NIHR Leeds Biomedical Research Centre, Chapel Allerton Hospital, Leeds, UK.
| |
Collapse
|
34
|
Shao Z, Wang B, Gao H, Zhang S. Microenvironmental interference with intra-articular stem cell regeneration influences the onset and progression of arthritis. Front Genet 2024; 15:1380696. [PMID: 38841721 PMCID: PMC11150611 DOI: 10.3389/fgene.2024.1380696] [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: 02/02/2024] [Accepted: 04/30/2024] [Indexed: 06/07/2024] Open
Abstract
Studies have indicated that the preservation of joint health and the facilitation of damage recovery are predominantly contingent upon the joint's microenvironment, including cell-cell interactions, the extracellular matrix's composition, and the existence of local growth factors. Mesenchymal stem cells (MSCs), which possess the capacity to self-renew and specialize in many directions, respond to cues from the microenvironment, and aid in the regeneration of bone and cartilage, are crucial to this process. Changes in the microenvironment (such as an increase in inflammatory mediators or the breakdown of the extracellular matrix) in the pathological context of arthritis might interfere with stem cell activation and reduce their ability to regenerate. This paper investigates the potential role of joint microenvironmental variables in promoting or inhibiting the development of arthritis by influencing stem cells' ability to regenerate. The present status of research on stem cell activity in the joint microenvironment is also outlined, and potential directions for developing new treatments for arthritis that make use of these intervention techniques to boost stem cell regenerative potential through altering the intra-articular environment are also investigated. This review's objectives are to investigate these processes, offer fresh perspectives, and offer a solid scientific foundation for the creation of arthritic treatment plans in the future.
Collapse
Affiliation(s)
| | | | | | - Shenqi Zhang
- Department of Joint and Sports Medicine, Zaozhuang Municipal Hospital Affiliated to Jining Medical University, Zaozhuang, Shandong, China
| |
Collapse
|
35
|
Liu X, Astudillo Potes MD, Dashtdar B, Schreiber AC, Tilton M, Li L, Elder BD, Lu L. 3D Stem Cell Spheroids with 2D Hetero-Nanostructures for In Vivo Osteogenic and Immunologic Modulated Bone Repair. Adv Healthc Mater 2024; 13:e2303772. [PMID: 38271276 PMCID: PMC11404522 DOI: 10.1002/adhm.202303772] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Indexed: 01/27/2024]
Abstract
3D stem cell spheroids have immense potential for various tissue engineering applications. However, current spheroid fabrication techniques encounter cell viability issues due to limited oxygen access for cells trapped within the core, as well as nonspecific differentiation issues due to the complicated environment following transplantation. In this study, functional 3D spheroids are developed using mesenchymal stem cells with 2D hetero-nanostructures (HNSs) composed of single-stranded DNA (ssDNA) binding carbon nanotubes (sdCNTs) and gelatin-bind black phosphorus nanosheets (gBPNSs). An osteogenic molecule, dexamethasone (DEX), is further loaded to fabricate an sdCNTgBP-DEX HNS. This approach aims to establish a multifunctional cell-inductive 3D spheroid with improved oxygen transportation through hollow nanotubes, stimulated stem cell growth by phosphate ions supplied from BP oxidation, in situ immunoregulation, and osteogenesis induction by DEX molecules after implantation. Initial transplantation of the 3D spheroids in rat calvarial bone defect shows in vivo macrophage shifts to an M2 phenotype, leading to a pro-healing microenvironment for regeneration. Prolonged implantation demonstrates outstanding in vivo neovascularization, osteointegration, and new bone regeneration. Therefore, these engineered 3D spheroids hold great promise for bone repair as they allow for stem cell delivery and provide immunoregulative and osteogenic signals within an all-in-one construct.
Collapse
Affiliation(s)
- Xifeng Liu
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN 55905, USA
| | - Maria D. Astudillo Potes
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN 55905, USA
| | - Babak Dashtdar
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN 55905, USA
| | - Areonna C. Schreiber
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN 55905, USA
| | - Maryam Tilton
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN 55905, USA
| | - Linli Li
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN 55905, USA
| | - Benjamin D. Elder
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN 55905, USA
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN 55905, USA
| | - Lichun Lu
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
36
|
Rajput SN, Naeem BK, Ali A, Salim A, Khan I. Expansion of human umbilical cord derived mesenchymal stem cells in regenerative medicine. World J Stem Cells 2024; 16:410-433. [PMID: 38690517 PMCID: PMC11056638 DOI: 10.4252/wjsc.v16.i4.410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 02/01/2024] [Accepted: 03/18/2024] [Indexed: 04/25/2024] Open
Abstract
BACKGROUND Stem cells are undifferentiated cells that possess the potential for self-renewal with the capacity to differentiate into multiple lineages. In humans, their limited numbers pose a challenge in fulfilling the necessary demands for the regeneration and repair of damaged tissues or organs. Studies suggested that mesenchymal stem cells (MSCs), necessary for repair and regeneration via transplantation, require doses ranging from 10 to 400 million cells. Furthermore, the limited expansion of MSCs restricts their therapeutic application. AIM To optimize a novel protocol to achieve qualitative and quantitative expansion of MSCs to reach the targeted number of cells for cellular transplantation and minimize the limitations in stem cell therapy protocols. METHODS Human umbilical cord (hUC) tissue derived MSCs were obtained and re-cultured. These cultured cells were subjected to the following evaluation procedures: Immunophenotyping, immunocytochemical staining, trilineage differentiation, population doubling time and number, gene expression markers for proliferation, cell cycle progression, senescence-associated β-galactosidase assay, human telomerase reverse transcriptase (hTERT) expression, mycoplasma, cytomegalovirus and endotoxin detection. RESULTS Analysis of pluripotent gene markers Oct4, Sox2, and Nanog in recultured hUC-MSC revealed no significant differences. The immunophenotypic markers CD90, CD73, CD105, CD44, vimentin, CD29, Stro-1, and Lin28 were positively expressed by these recultured expanded MSCs, and were found negative for CD34, CD11b, CD19, CD45, and HLA-DR. The recultured hUC-MSC population continued to expand through passage 15. Proliferative gene expression of Pax6, BMP2, and TGFb1 showed no significant variation between recultured hUC-MSC groups. Nevertheless, a significant increase (P < 0.001) in the mitotic phase of the cell cycle was observed in recultured hUC-MSCs. Cellular senescence markers (hTERT expression and β-galactosidase activity) did not show any negative effect on recultured hUC-MSCs. Additionally, quality control assessments consistently confirmed the absence of mycoplasma, cytomegalovirus, and endotoxin contamination. CONCLUSION This study proposes the development of a novel protocol for efficiently expanding stem cell population. This would address the growing demand for larger stem cell doses needed for cellular transplantation and will significantly improve the feasibility of stem cell based therapies.
Collapse
Affiliation(s)
- Shafiqa Naeem Rajput
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Sindh, Pakistan
| | - Bushra Kiran Naeem
- Surgical Unit 4, Dr. Ruth KM Pfau Civil Hospital, Karachi 74400, Pakistan
| | - Anwar Ali
- Department of Physiology, University of Karachi, Karachi 75270, Pakistan
| | - Asmat Salim
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Sindh, Pakistan
| | - Irfan Khan
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Sindh, Pakistan
- Center for Regenerative Medicine and Stem Cells Research, and Department of Ophthalmology and Visual Sciences, The Aga Khan University, Karachi 74800, Sindh, Pakistan.
| |
Collapse
|
37
|
Tamaño-Machiavello M, Carvalho E, Correia D, Cordón L, Lanceros-Méndez S, Sempere A, Sabater i Serra R, Ribelles JG. Osteogenic differentiation of human mesenchymal stem cells on electroactive substrates. Heliyon 2024; 10:e28880. [PMID: 38601667 PMCID: PMC11004758 DOI: 10.1016/j.heliyon.2024.e28880] [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: 10/19/2023] [Revised: 03/26/2024] [Accepted: 03/26/2024] [Indexed: 04/12/2024] Open
Abstract
This study investigates the effect of electroactivity and electrical charge distribution on the biological response of human bone marrow stem cells (hBMSCs) cultured in monolayer on flat poly(vinylidene fluoride), PVDF, substrates. Differences in cell behaviour, including proliferation, expression of multipotency markers CD90, CD105 and CD73, and expression of genes characteristic of different mesenchymal lineages, were observed both during expansion in basal medium before reaching confluence and in confluent cultures in osteogenic induction medium. The crystallisation of PVDF in the electrically neutral α-phase or in the electroactive phase β, both unpoled and poled, has been found to have an important influence on the biological response. In addition, the presence of a permanent positive or negative surface electrical charge distribution in phase β substrates has also shown a significant effect on cell behaviour.
Collapse
Affiliation(s)
- M.N. Tamaño-Machiavello
- Centre for Biomaterials and Tissue Engineering, CBIT, Universitat Politècnica de València, 46022 València, Spain
| | - E.O. Carvalho
- Centre of Physics, Universidade do Minho, 4710-057, Braga, Portugal
| | - D. Correia
- Centre of Chemistry, University of Minho, 4710-057, Braga, Portugal
| | - L. Cordón
- Hematology Research Group, Instituto de Investigación Sanitaria La Fe, València, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto Carlos III, Madrid, Spain
| | - S. Lanceros-Méndez
- Centre of Physics, Universidade do Minho, 4710-057, Braga, Portugal
- BCMaterials, Basque Center for Materials, Applications and Nanostructures, UPV/EHU Science Park, 48940, Leioa, Spain
- IKERBASQUE, Basque Foundation for Science, 48009, Bilbao, Spain
| | - A. Sempere
- Hematology Research Group, Instituto de Investigación Sanitaria La Fe, València, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto Carlos III, Madrid, Spain
- Hematology Department, Hospital Universitario y Politécnico La Fe, València, Spain
| | - R. Sabater i Serra
- Centre for Biomaterials and Tissue Engineering, CBIT, Universitat Politècnica de València, 46022 València, Spain
- CIBER de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, Spain
| | - J.L. Gómez Ribelles
- Centre for Biomaterials and Tissue Engineering, CBIT, Universitat Politècnica de València, 46022 València, Spain
- CIBER de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, Spain
| |
Collapse
|
38
|
Aggarwal A, Nath D, Pal A, Sah MK. Harnessing potential of avian eggshell membrane derived collagen hydrolysate for bone tissue regeneration. Mol Biol Rep 2024; 51:482. [PMID: 38578512 DOI: 10.1007/s11033-024-09394-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 02/28/2024] [Indexed: 04/06/2024]
Abstract
BACKGROUND Natural bone grafts are the highly preferred materials for restoring the lost bone, while being constrained of donor availability and risk of disease transmission. As a result, tissue engineering is emerging as an efficacious and competitive technique for bone repair. Bone tissue engineering (TE) scaffolds to support bone regeneration and devoid of aforesaid limitations are being vastly explored and among these the avian eggshell membrane has drawn attention for TE owing to its low immunogenicity, similarity with the extracellular matrix, and easy availability. METHODOLOGY AND RESULTS In this study, the development of bone ingrowth support system from avian eggshell membrane derived collagen hydrolysates (Col-h) is reported. The hydrolysate, cross-linked with glutaraldehyde, was developed into hydrogels with poly-(vinyl alcohol) (PVA) by freeze-thawing and further characterized with ATR-FTIR, XRD, FESEM. The biodegradability, swelling, mechanical, anti-microbial, and biocompatibility evaluation were performed further for the suitability in bone regeneration. The presence of amide I, amide III, and -OH functional groups at 1639 cm- 1,1264 cm- 1, and 3308 cm- 1 respectively and broad peak between 16°-21° (2θ) in XRD data reinstated the composition and form. CONCLUSIONS The maximum ratio of Col-h/PVA that produced well defined hydrogels was 50:50. Though all the hydrogel matrices alluded towards their competitive attributes and applicability towards restorative bone repair, the hydrogel with 40:60 ratios showed better mechanical strength and cell proliferation than its counterparts. The prominent E. coli growth inhibition by the hydrogel matrices was also observed, along with excellent biocompatibility with MG-63 osteoblasts. The findings indicate strongly the promising application of avian eggshell-derived Col-h in supporting bone regeneration.
Collapse
Affiliation(s)
- Aakriti Aggarwal
- Department of Biotechnology, Dr. B. R. Ambedkar National Institute of Technology, Jalandhar, Punjab, 1440081, India
| | - Debasish Nath
- Chemical Biology Unit, Institute of Nanoscience and Technology, Sector 81, Mohali, Punjab, 140306, India
| | - Asish Pal
- Chemical Biology Unit, Institute of Nanoscience and Technology, Sector 81, Mohali, Punjab, 140306, India
| | - Mahesh Kumar Sah
- Department of Biotechnology, Dr. B. R. Ambedkar National Institute of Technology, Jalandhar, Punjab, 1440081, India.
| |
Collapse
|
39
|
Sheppard AJ, Delgado K, Barfield AM, Xu Q, Massey PA, Dong Y, Barton RS. Rapamycin Inhibits Senescence and Improves Immunomodulatory Function of Mesenchymal Stem Cells Through IL-8 and TGF-β Signaling. Stem Cell Rev Rep 2024; 20:816-826. [PMID: 38340274 PMCID: PMC10984889 DOI: 10.1007/s12015-024-10682-x] [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] [Accepted: 01/16/2024] [Indexed: 02/12/2024]
Abstract
Mesenchymal stromal cells (MSCs) grown in high-density monolayers (sheets) are promising vehicles for numerous bioengineering applications. When MSC sheets are maintained in prolonged cultures, they undergo rapid senescence, limiting their downstream efficacy. Although rapamycin is a potential agent that can inhibit senescence in cell cultures, no study has investigated rapamycin's effect on MSCs grown in high-density culture and its effect on downstream target gene expression. In this study, placental-derived MSCs (PMSCs) were seeded at high density to generate PMSC sheets in 24 hours and were then treated with rapamycin or vehicle for up to 7 days. Autophagy activity, cell senescence and apoptosis, cell size and granularity, and senescence-associated cytokines (IL-6 and IL-8) were analyzed. Differential response in gene expression were assessed via microarray analysis. Rapamycin significantly increased PMSC sheet autophagy activity, inhibited cellular senescence, decreased cell size and granularity at all timepoints. Rapamycin also significantly decreased the number of cells in late apoptosis at day 7 of sheet culture, as well as caspase 3/7 activity at all timepoints. Notably, while rapamycin decreased IL-6 secretion, increased IL-8 levels were observed at all timepoints. Microarray analysis further confirmed the upregulation of IL-8 transcription, as well as provided a list of 396 genes with 2-fold differential expression, where transforming growth factor-β (TGF-β) signaling were identified as important upregulated pathways. Rapamycin both decreased senescence and has an immunomodulatory action of PMSCs grown in sheet culture, which will likely improve the chemotaxis of pro-healing cells to sites of tissue repair in future bioengineering applications.
Collapse
Affiliation(s)
- Aaron J Sheppard
- School of Medicine, LSU Health Shreveport, Shreveport, LA, USA
- Department of Orthopedic Surgery, LSU Health Shreveport, Shreveport, LA, USA
| | - Kristin Delgado
- School of Medicine, LSU Health Shreveport, Shreveport, LA, USA
| | | | - Qinqin Xu
- Department of Orthopedic Surgery, LSU Health Shreveport, Shreveport, LA, USA
| | - Patrick A Massey
- Department of Orthopedic Surgery, LSU Health Shreveport, Shreveport, LA, USA
| | - Yufeng Dong
- Department of Orthopedic Surgery, LSU Health Shreveport, Shreveport, LA, USA.
| | - Richard S Barton
- Department of Orthopedic Surgery, LSU Health Shreveport, Shreveport, LA, USA
| |
Collapse
|
40
|
Muraev AA, Manukyan GG, Salekh KM, Bonartsev AP, Volkov AV. Magnetic field application in bone tissue regeneration: issue current status and prospects for method development. RUDN JOURNAL OF MEDICINE 2024; 28:9-22. [DOI: 10.22363/2313-0245-2024-28-1-9-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2025] Open
Abstract
Relevance. Magnets have long been used to treat various diseases, especially in inflammatory processes. According to existing historical data, magnetotherapy was already used in ancient times by the Chinese, Egyptians and Greeks. Different magnetic field strengths affect cells in different ways, with medium-strength magnetic fields being the most widely used. The review presents a brief history and current state of the issue of using a magnetic field in bone tissue regeneration. Modern knowledge about the mechanisms of physiological and reparative regeneration, restoration of bone tissue is clarified, and modern areas of bone tissue engineering are considered, taking into account the characteristics of microcirculation and the effect of a magnetic field on the physiology of bone tissue and reparative regeneration. One of the key findings of the review is that the magnetic field improves bone tissue repair by influencing the metabolic behavior of cells. Studies show that magnetotherapy promotes the activation of cellular processes, accelerates the formation of new bone tissue and improves its quality. It is also noted that the magnetic field has a positive effect on microcirculation, improving the blood supply to tissues and facilitating a better supply of nutrients to the site of injury. This contributes to faster wound healing and early rehabilitation of patients. Conclusion. Magnetotherapy is one of the effective physical and rehabilitation methods of treatment that will become increasingly important in modern medicine. However, further research is needed to better understand the mechanisms of action of a magnetic field on bone tissue and to determine the optimal parameters for its application.
Collapse
|
41
|
Tivig I, Vallet L, Moisescu MG, Fernandes R, Andre FM, Mir LM, Savopol T. Early differentiation of mesenchymal stem cells is reflected in their dielectrophoretic behavior. Sci Rep 2024; 14:4330. [PMID: 38383752 PMCID: PMC10881469 DOI: 10.1038/s41598-024-54350-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 02/12/2024] [Indexed: 02/23/2024] Open
Abstract
The therapeutic use of mesenchymal stem cells (MSCs) becomes more and more important due to their potential for cell replacement procedures as well as due to their immunomodulatory properties. However, protocols for MSCs differentiation can be lengthy and may result in incomplete or asynchronous differentiation. To ensure homogeneous populations for therapeutic purposes, it is crucial to develop protocols for separation of the different cell types after differentiation. In this article we show that, when MSCs start to differentiate towards adipogenic or osteogenic progenies, their dielectrophoretic behavior changes. The values of cell electric parameters which can be obtained by dielectrophoretic measurements (membrane permittivity, conductivity, and cytoplasm conductivity) change before the morphological features of differentiation become microscopically visible. We further demonstrate, by simulation, that these electric modifications make possible to separate cells in their early stages of differentiation by using the dielectrophoretic separation technique. A label free method which allows obtaining cultures of homogenously differentiated cells is thus offered.
Collapse
Grants
- PN-III-P2-2.1-PED-2021, grant no. 596PED/2022 Romanian Executive Agency for Higher Education, Research, Development, and Innovation Funding
- PN-III-P2-2.1-PED-2021, grant no. 596PED/2022 Romanian Executive Agency for Higher Education, Research, Development, and Innovation Funding
- PN-III-P2-2.1-PED-2021, grant no. 596PED/2022 Romanian Executive Agency for Higher Education, Research, Development, and Innovation Funding
- PN-III-P3-3.1-PM-RO-FR-2019, grant no. 11BM/2019 Romania-France cooperation program Hubert Curien-Brancusi
- PN-III-P3-3.1-PM-RO-FR-2019, grant no. 11BM/2019 Romania-France cooperation program Hubert Curien-Brancusi
- PN-III-P3-3.1-PM-RO-FR-2019, grant no. 11BM/2019 Romania-France cooperation program Hubert Curien-Brancusi
- PN-III-P3-3.1-PM-RO-FR-2019, grant no. 11BM/2019 Romania-France cooperation program Hubert Curien-Brancusi
- FET-OPEN H2020, grant no. 964562 Horizon 2020
- FET-OPEN H2020, grant no. 964562 Horizon 2020
- FET-OPEN H2020, grant no. 964562 Horizon 2020
Collapse
Affiliation(s)
- Ioan Tivig
- Biophysics and Cellular Biotechnology Department, Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 8 Eroii Sanitari Blvd., 050474, Bucharest, Romania
- Excellence Center for Research in Biophysics and Cellular Biotechnology, Carol Davila University of Medicine and Pharmacy, 050474, Bucharest, Romania
| | - Leslie Vallet
- METSY UMR 9018, Université Paris-Saclay, CNRS and Gustave Roussy, 94805, Villejuif, France
| | - Mihaela G Moisescu
- Biophysics and Cellular Biotechnology Department, Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 8 Eroii Sanitari Blvd., 050474, Bucharest, Romania.
- Excellence Center for Research in Biophysics and Cellular Biotechnology, Carol Davila University of Medicine and Pharmacy, 050474, Bucharest, Romania.
| | - Romain Fernandes
- METSY UMR 9018, Université Paris-Saclay, CNRS and Gustave Roussy, 94805, Villejuif, France
| | - Franck M Andre
- METSY UMR 9018, Université Paris-Saclay, CNRS and Gustave Roussy, 94805, Villejuif, France
| | - Lluis M Mir
- METSY UMR 9018, Université Paris-Saclay, CNRS and Gustave Roussy, 94805, Villejuif, France
| | - Tudor Savopol
- Biophysics and Cellular Biotechnology Department, Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 8 Eroii Sanitari Blvd., 050474, Bucharest, Romania
- Excellence Center for Research in Biophysics and Cellular Biotechnology, Carol Davila University of Medicine and Pharmacy, 050474, Bucharest, Romania
| |
Collapse
|
42
|
Park JS, Kim DY, Hong HS. FGF2/HGF priming facilitates adipose-derived stem cell-mediated bone formation in osteoporotic defects. Heliyon 2024; 10:e24554. [PMID: 38304814 PMCID: PMC10831751 DOI: 10.1016/j.heliyon.2024.e24554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 12/14/2023] [Accepted: 01/10/2024] [Indexed: 02/03/2024] Open
Abstract
Aims The activity of adipose-derived stem cells (ADSCs) is susceptible to the physiological conditions of the donor. Therefore, employing ADSCs from donors of advanced age or with diseases for cell therapy necessitates a strategy to enhance therapeutic efficacy before transplantation. This study aims to investigate the impact of supplementing Fibroblast Growth Factor 2 (FGF2) and Hepatocyte Growth Factor (HGF) on ADSC-mediated osteogenesis under osteoporotic conditions and to explore the underlying mechanisms of action. Main methods Adipose-derived stem cells (ADSCs) obtained from ovariectomized (OVX) rats were cultured ex vivo. These cells were cultured in an osteogenic medium supplemented with FGF2 and HGF and subsequently autologously transplanted into osteoporotic femur defects using Hydroxyapatite-Tricalcium Phosphate. The assessment of bone formation was conducted four weeks post-transplantation. Key findings Osteoporosis detrimentally affects the viability and osteogenic differentiation potential of ADSCs, often accompanied by a deficiency in FGF2 and HGF signaling. However, priming with FGF2 and HGF facilitated the formation of immature osteoblasts from OVX ADSCs in vitro, promoting the expression of osteoblastogenic proteins, including Runx-2, osterix, and ALP, during the early phase of osteogenesis. Furthermore, FGF2/HGF priming augmented the levels of VEGF and SDF-1α in the microenvironment of OVX ADSCs under osteogenic induction. Importantly, transplantation of OVX ADSCs primed with FGF2/HGF for 6 days significantly enhanced bone formation compared to non-primed cells. The success of bone regeneration was confirmed by the expression of type-1 collagen and osteocalcin in the bone tissue of the deficient area. Significance Our findings corroborate that priming with FGF2/HGF can improve the differentiation potential of ADSCs. This could be applied in autologous stem cell therapy for skeletal disease in the geriatric population.
Collapse
Affiliation(s)
- Jeong Seop Park
- Department of Biomedical Science and Technology, Graduate School, Kyung Hee University, Seoul, 02447, South Korea
| | - Do Young Kim
- Department of Biomedical Science and Technology, Graduate School, Kyung Hee University, Seoul, 02447, South Korea
| | - Hyun Sook Hong
- Department of Biomedical Science and Technology, Graduate School, Kyung Hee University, Seoul, 02447, South Korea
- East-West Medical Research Institute, Kyung Hee University, Seoul, 02447, South Korea
- Kyung Hee Institute of Regenerative Medicine (KIRM), Medical Science Research Institute, Kyung Hee University Medical Center, Seoul, 02447, South Korea
| |
Collapse
|
43
|
Tao X, Wang J, Liu B, Cheng P, Mu D, Du H, Niu B. Plasticity and crosstalk of mesenchymal stem cells and macrophages in immunomodulation in sepsis. Front Immunol 2024; 15:1338744. [PMID: 38352879 PMCID: PMC10861706 DOI: 10.3389/fimmu.2024.1338744] [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: 11/15/2023] [Accepted: 01/10/2024] [Indexed: 02/16/2024] Open
Abstract
Sepsis is a multisystem disease characterized by dysregulation of the host immune response to infection. Immune response kinetics play a crucial role in the pathogenesis and progression of sepsis. Macrophages, which are known for their heterogeneity and plasticity, actively participate in the immune response during sepsis. These cells are influenced by the ever-changing immune microenvironment and exhibit two-sided immune regulation. Recently, the immunomodulatory function of mesenchymal stem cells (MSCs) in sepsis has garnered significant attention. The immune microenvironment can profoundly impact MSCs, prompting them to exhibit dual immunomodulatory functions akin to a double-edged sword. This discovery holds great importance for understanding sepsis progression and devising effective treatment strategies. Importantly, there is a close interrelationship between macrophages and MSCs, characterized by the fact that during sepsis, these two cell types interact and cooperate to regulate inflammatory processes. This review summarizes the plasticity of macrophages and MSCs within the immune microenvironment during sepsis, as well as the intricate crosstalk between them. This remains an important concern for the future use of these cells for immunomodulatory treatments in the clinic.
Collapse
Affiliation(s)
- Xingyu Tao
- Department of Critical Care Medicine, Chongqing Key Laboratory of Emergency Medicine, School of Medicine, Chongqing University Central Hospital, Chongqing University, Chongqing, China
| | - Jialian Wang
- Department of Critical Care Medicine, Chongqing Key Laboratory of Emergency Medicine, School of Medicine, Chongqing University Central Hospital, Chongqing University, Chongqing, China
| | - Bin Liu
- Department of Critical Care Medicine, Chongqing Key Laboratory of Emergency Medicine, School of Medicine, Chongqing University Central Hospital, Chongqing University, Chongqing, China
| | - Peifeng Cheng
- Department of Critical Care Medicine, Chongqing Key Laboratory of Emergency Medicine, School of Medicine, Chongqing University Central Hospital, Chongqing University, Chongqing, China
| | - Dan Mu
- Department of Critical Care Medicine, Chongqing Key Laboratory of Emergency Medicine, School of Medicine, Chongqing University Central Hospital, Chongqing University, Chongqing, China
| | - Huimin Du
- Department of Oncology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Bailin Niu
- Department of Critical Care Medicine, Chongqing Key Laboratory of Emergency Medicine, School of Medicine, Chongqing University Central Hospital, Chongqing University, Chongqing, China
- Department of Surgery, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, United States
| |
Collapse
|
44
|
Wang J, Jing J, Zhou C, Fan Y. Emerging roles of exosomes in oral diseases progression. Int J Oral Sci 2024; 16:4. [PMID: 38221571 PMCID: PMC10788352 DOI: 10.1038/s41368-023-00274-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 12/11/2023] [Accepted: 12/26/2023] [Indexed: 01/16/2024] Open
Abstract
Oral diseases, such as periodontitis, salivary gland diseases, and oral cancers, significantly challenge health conditions due to their detrimental effects on patient's digestive functions, pronunciation, and esthetic demands. Delayed diagnosis and non-targeted treatment profoundly influence patients' prognosis and quality of life. The exploration of innovative approaches for early detection and precise treatment represents a promising frontier in oral medicine. Exosomes, which are characterized as nanometer-sized extracellular vesicles, are secreted by virtually all types of cells. As the research continues, the complex roles of these intracellular-derived extracellular vesicles in biological processes have gradually unfolded. Exosomes have attracted attention as valuable diagnostic and therapeutic tools for their ability to transfer abundant biological cargos and their intricate involvement in multiple cellular functions. In this review, we provide an overview of the recent applications of exosomes within the field of oral diseases, focusing on inflammation-related bone diseases and oral squamous cell carcinomas. We characterize the exosome alterations and demonstrate their potential applications as biomarkers for early diagnosis, highlighting their roles as indicators in multiple oral diseases. We also summarize the promising applications of exosomes in targeted therapy and proposed future directions for the use of exosomes in clinical treatment.
Collapse
Affiliation(s)
- Jiayi Wang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Junjun Jing
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Chenchen Zhou
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yi Fan
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China.
| |
Collapse
|
45
|
Salamanna F, Tedesco G, Sartori M, Griffoni C, Spinnato P, Romeo P, Ghermandi R, Fini M, Giavaresi G, Gasbarrini A, Barbanti Brodano G. Safety and efficacy of autologous bone marrow clot as a multifunctional bioscaffold for instrumental posterior lumbar fusion: a 1-year follow-up pilot study. Front Endocrinol (Lausanne) 2024; 14:1245344. [PMID: 38260131 PMCID: PMC10801235 DOI: 10.3389/fendo.2023.1245344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 12/12/2023] [Indexed: 01/24/2024] Open
Abstract
Background Bone marrow aspirate (BMA), when combined with graft substitutes, has long been introduced as a promising alternative to iliac crest bone graft in spinal fusion. However, the use of BMA is limited by the absence of a standardized procedure, a structural texture, and the potential for diffusion away from the implant site. Recently, the potential use of a new formulation of BMA, named BMA clot, has been preclinically described. In this report, we present the results of a prospective pilot clinical study aimed at evaluating the safety and efficacy of autologous vertebral BMA (vBMA) clot as a three-dimensional and multifunctional bioscaffold in instrumented posterior lumbar fusion. Methods Ten consecutive patients with an indication of multilevel (≤5) posterior spinal fusion due to lumbar spine degenerative diseases were included in the study and treated with vBMA. Clinical outcomes were assessed using the Visual Analog Scale (VAS), Oswestry Disability Index (ODI), and EuroQoL-5L (EQ-5L) preoperatively and at 3 months and 12 months after spinal fusion. Bone fusion quality was evaluated at the 12-month follow-up using the Brantigan classification on radiography (XR) imaging. Bone density was measured on computed tomography (CT) scans at 6 and 12 months of follow-up visits at the intervertebral arches and intervertebral joint areas and expressed in Hounsfield unit (HU). Results The results indicate a successful posterolateral fusion rate of approximately 100% (considering levels with C, D, and E grades according to the Brantigan classification) at the 12-month follow-up, along with an increase in bone density from 6 to 12 months of follow-up. An improvement in the quality of life and health status following surgery, as assessed by clinical scores (ODI, VAS, and EQ-5L), was also observed as early as 3 months postsurgery. No adverse events related to the vBMA clot were reported. Conclusion This prospective pilot study demonstrates the effectiveness and safety profile of vBMA clot as an advanced bioscaffold capable of achieving posterior lumbar fusion in the treatment of degenerative spine diseases. This lays the groundwork for a larger randomized clinical study.
Collapse
Affiliation(s)
- Francesca Salamanna
- Surgical Sciences and Technologies, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Giuseppe Tedesco
- Spine Surgery Unit, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Maria Sartori
- Surgical Sciences and Technologies, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | | | - Paolo Spinnato
- Diagnostic and Interventional Radiology, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Paolo Romeo
- Diagnostic and Interventional Radiology, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | | | - Milena Fini
- Scientific Direction, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Gianluca Giavaresi
- Surgical Sciences and Technologies, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | | | | |
Collapse
|
46
|
Gu Y, Song Y, Pan Y, Liu J. The essential roles of m 6A modification in osteogenesis and common bone diseases. Genes Dis 2024; 11:335-345. [PMID: 37588215 PMCID: PMC10425797 DOI: 10.1016/j.gendis.2023.01.032] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 01/30/2023] [Indexed: 03/30/2023] Open
Abstract
N6-methyladenosine (m6A) is the most prevalent modification in the eukaryotic transcriptome and has a wide range of functions in coding and noncoding RNAs. It affects the fate of the modified RNA, including its stability, splicing, and translation, and plays an important role in post-transcriptional regulation. Bones play a key role in supporting and protecting muscles and other organs, facilitating the movement of the organism, ensuring blood production, etc. Bone diseases such as osteoarthritis, osteoporosis, and bone tumors are serious public health problems. The processes of bone development and osteogenic differentiation require the precise regulation of gene expression through epigenetic mechanisms including histone, DNA, and RNA modifications. As a reversible dynamic epigenetic mark, m6A modifications affect nearly every important biological process, cellular component, and molecular function, including skeletal development and homeostasis. In recent years, studies have shown that m6A modification is involved in osteogenesis and bone-related diseases. In this review, we summarized the proteins involved in RNA m6A modification and the latest progress in elucidating the regulatory role of m6A modification in bone formation and stem cell directional differentiation. We also discussed the pathological roles and potential molecular mechanisms of m6A modification in bone-related diseases like osteoporosis and osteosarcoma and suggested potential areas for new strategies that could be used to prevent or treat bone defects and bone diseases.
Collapse
Affiliation(s)
- Yuxi Gu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yidan Song
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yihua Pan
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Jun Liu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| |
Collapse
|
47
|
Tan X, Zhang M, Tu B. Evaluation of bioactive extract nanoparticles on pulp stem cell behavior relevant to dental care using chemical composition of gelatin-Arabian gum nano polymer. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2024; 27:975-984. [PMID: 38911239 PMCID: PMC11193506 DOI: 10.22038/ijbms.2024.76467.16548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Accepted: 02/18/2024] [Indexed: 06/25/2024]
Abstract
Objectives This study aimed to investigate the impact of bioactive plant extracts on the proliferation and migration of dental pulp stem cells (DPSCs) and their potential implications for dental care, focusing on the nurse-caring aspect. Materials and Methods TDPSCs were cultured on gelatin polymer scaffolds mimicking the extracellular matrix (ECM) environment. Bioactive plant extracts with antibacterial, anti-inflammatory, and anti-oxidant properties were incorporated into the gelatin polymer at concentrations ranging from 0.1% to 2.0%. Proliferation and migration assays were performed, considering nurse-caring practices during the experiments. Results Treatment with specific bioactive plant extracts significantly enhanced DPSC proliferation, showing a 2.5-fold increase compared to the control groups. The migration assay revealed a substantial increase in cell migration distance, with treated cells covering an average distance of 400-500 μm compared to 220-260 μm in the control group. Treated cells also exhibited improved viability and metabolic activity, with a 30% increase in cell viability and a 10-20% increase in metabolic activity compared to the control group. Conclusion This study demonstrates that bioactive plant extracts have the potential to enhance DPSC proliferation, migration, viability, and metabolic activity. These findings support the use of these extracts in dental care, benefiting from the nurse-caring practices.
Collapse
Affiliation(s)
- Xiaoni Tan
- Department of Endodontics, Changsha Stomatological Hospital, No.389, Youyi Road, Tianxin District, Changsha 410008, Hunan Province, China
| | - Moli Zhang
- Department of Endodontics, Changsha Stomatological Hospital, No.389, Youyi Road, Tianxin District, Changsha 410008, Hunan Province, China
| | - BiBo Tu
- Department of Endodontics, Changsha Stomatological Hospital, No.389, Youyi Road, Tianxin District, Changsha 410008, Hunan Province, China
| |
Collapse
|
48
|
Karima G, Kim HD. Unlocking the regenerative key: Targeting stem cell factors for bone renewal. J Tissue Eng 2024; 15:20417314241287491. [PMID: 39479284 PMCID: PMC11523181 DOI: 10.1177/20417314241287491] [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: 06/22/2024] [Accepted: 09/12/2024] [Indexed: 11/02/2024] Open
Abstract
Stem cell factors (SCFs) are pivotal factors existing in both soluble and membrane-bound forms, expressed by endothelial cells (ECs) and fibroblasts throughout the body. These factors enhance cell growth, viability, and migration in multipotent cell lineages. The preferential expression of SCF by arteriolar ECs indicates that arterioles create a unique microenvironment tailored to hematopoietic stem cells (HSCs). Insufficiency of SCF within bone marrow (BM)-derived adipose tissue results in decreased their overall cellularity, affecting HSCs and their immediate progenitors critical for generating diverse blood cells and maintaining the hematopoietic microenvironment. SCF deficiency disrupts BM function, impacting the production and differentiation of HSCs. Additionally, deleting SCF from adipocytes reduces lipogenesis, highlighting the crucial role of SCF/c-kit signaling in controlling lipid accumulation. This review elucidates the sources, roles, mechanisms, and molecular strategies of SCF in bone renewal, offering a comprehensive overview of recent advancements, challenges, and future directions for leveraging SCF as a key agent in regenerative medicine.
Collapse
Affiliation(s)
- Gul Karima
- Department of Polymer Science and Engineering, Korea National University of Transportation, Chungju, Republic of Korea
| | - Hwan D. Kim
- Department of Polymer Science and Engineering, Korea National University of Transportation, Chungju, Republic of Korea
- Department of IT Convergence (Brain Korea Plus 21), Korea National University of Transportation, Chungju, Republic of Korea
- Department of Biomedical Engineering, Korea National University of Transportation, Chungju, Republic of Korea
| |
Collapse
|
49
|
Li T, Peng Z, Lv Q, Li L, Zhang C, Pang L, Zhang C, Li Y, Chen Y, Tang X. SLS 3D Printing To Fabricate Poly(vinyl alcohol)/Hydroxyapatite Bioactive Composite Porous Scaffolds and Their Bone Defect Repair Property. ACS Biomater Sci Eng 2023; 9:6734-6744. [PMID: 37939039 DOI: 10.1021/acsbiomaterials.3c01014] [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] [Indexed: 11/10/2023]
Abstract
Poly(vinyl alcohol) (PVA) exhibits a wide range of potential applications in the biomedical field due to its favorable mechanical properties and biocompatibility. However, few studies have been carried out on selective laser sintering (SLS) of PVA due to its poor thermal processability. In this study, in order to impart PVA powder the excellent thermal processability, the molecular complexation technology was performed to destroy the strong hydrogen bonds in PVA and thus significantly reduced the PVA melting point and crystallinity to 190.9 °C and 27.9%, respectively. The modified PVA (MPVA) was then compounded with hydroxyapatite (HA) to prepare PVA/HA composite powders suitable for SLS 3D printing. The final SLS 3D-printed MPVA/HA composite porous scaffolds show high precision and interconnected pores with a porosity as high as 68.3%. The in vitro cell culture experiments revealed that the sintered composite scaffolds could significantly promote the adhesion and proliferation of osteoblasts and facilitate bone regeneration, and the quantitative real-time polymerase chain reaction results further demonstrate that the printed MPVA/20HA scaffold could significantly enhance the expression levels of both early osteogenic-specific marker of alkaline phosphatase stain and runt-related transcription factor 2. Meanwhile, in in vivo experiments, it is encouragingly found that the resultant MPVA/20HA SLS 3D-printed part has an obvious effect on promoting the growth of new bone tissue as well as a better bone regeneration capability. This work could provide a promising strategy for fabrication of PVA scaffolds through SLS 3D printing, exhibiting a great potential for clinical applications in bone tissue engineering.
Collapse
Affiliation(s)
- Tao Li
- Sports Medicine Center, West China Hospital, Sichuan University, Chengdu 610065, China
- Department of Orthopedics and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu 610065, China
| | - Zilin Peng
- The State Key Laboratory of Polymer Materials Engineering, Polymer Research Institute of Sichuan University, Chengdu 610065, China
| | - Qinniu Lv
- The State Key Laboratory of Polymer Materials Engineering, Polymer Research Institute of Sichuan University, Chengdu 610065, China
| | - Li Li
- The State Key Laboratory of Polymer Materials Engineering, Polymer Research Institute of Sichuan University, Chengdu 610065, China
| | - Chuhong Zhang
- The State Key Laboratory of Polymer Materials Engineering, Polymer Research Institute of Sichuan University, Chengdu 610065, China
| | - Long Pang
- Sports Medicine Center, West China Hospital, Sichuan University, Chengdu 610065, China
- Department of Orthopedics and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu 610065, China
| | - Chunsen Zhang
- Sports Medicine Center, West China Hospital, Sichuan University, Chengdu 610065, China
- Department of Orthopedics and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu 610065, China
| | - Yinghao Li
- Sports Medicine Center, West China Hospital, Sichuan University, Chengdu 610065, China
- Department of Orthopedics and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu 610065, China
| | - Yinghong Chen
- The State Key Laboratory of Polymer Materials Engineering, Polymer Research Institute of Sichuan University, Chengdu 610065, China
| | - Xin Tang
- Sports Medicine Center, West China Hospital, Sichuan University, Chengdu 610065, China
- Department of Orthopedics and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu 610065, China
| |
Collapse
|
50
|
Dibazar ZE, Zarei M, Mohammadikhah M, Oudah SK, Elyasi M, Kokabi H, Shahgolzari M, Asl LD, Azizy M. Crosslinking strategies for biomimetic hydrogels in bone tissue engineering. Biophys Rev 2023; 15:2027-2040. [PMID: 38192345 PMCID: PMC10771399 DOI: 10.1007/s12551-023-01141-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 09/03/2023] [Indexed: 01/10/2024] Open
Abstract
Bone tissue engineering has become a popular area of study for making biomimetic hydrogels to treat bone diseases. In this work, we looked at biocompatible hydrogels that can be injected into bone defects that require the smallest possible surgery. Mineral ions can be attached to polymer chains to make useful hydrogels that help bones heal faster. These ions are very important for the balance of the body. In the chemically-triggered sector, advanced hydrogels cross-linked by different molecular agents have many advantages, such as being selective, able to form gels, and having mechanical properties that can be modified. In addition, different photo-initiators can be used to make photo cross linkable hydrogels react quickly and moderately under certain light bands. Enzyme-triggered hydrogels are another type of hydrogel that can be used to repair bone tissue because they are biocompatible and gel quickly. We also look at some of the important factors mentioned above that could change how well bone tissue engineering works as a therapy. Finally, this review summarizes the problems that still need to be solved to make clinically relevant hydrogels.
Collapse
Affiliation(s)
- Zahra Ebrahimvand Dibazar
- Department of Oral and Maxillo Facial Medicine, Faculty of Dentistry, Tabriz Azad University of Medical Sciences, Tabriz, 5165687386 Iran
| | - Mahdi Zarei
- Student Research Committee, Tabriz university of medical sciences, Tabriz, Iran
| | - Meysam Mohammadikhah
- Department of Oral and Maxillofacial Surgery, School of Dentistry, Alborz University of Medical Sciences, Karaj, Iran
| | - Shamam Kareem Oudah
- College of Pharmacy, National University of Science and Technology, Dhi Qar, Iraq
| | - Milad Elyasi
- Otolaryngology department, Shahid Beheshti University of medical sciences, Tehran, Iran
| | - Hadi Kokabi
- Department of Periodontics, School of Dentistry, Hamadan University of Medical Sciences, Hamadan, 65175-4171 Iran
| | - Mehdi Shahgolzari
- Dental Research Center, Hamadan University of Medical Sciences, Hamadan, 65175-4171 Iran
| | - Leila Delnabi Asl
- Department of Internal Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mahdi Azizy
- Department of Oral and Maxillofacial Surgery, School of Dentistry, Alborz University of Medical Sciences, Karaj, Iran
| |
Collapse
|