1
|
Jin X, Lu Y, Fan Z. Exploring NamiRNA networks and time-series gene expression in osteogenic differentiation of adipose-derived stem cells. Ann Med 2025; 57:2478323. [PMID: 40100054 PMCID: PMC11921168 DOI: 10.1080/07853890.2025.2478323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 02/12/2025] [Accepted: 02/28/2025] [Indexed: 03/20/2025] Open
Abstract
BACKGROUND Adipose-derived stem cells (ADSCs) are a type of stem cell found in adipose tissue with the capacity to differentiate into multiple lineages, including osteoblasts. The differentiation of ADSCs into osteoblasts underlies osteogenic and pathological cellular basis in osteoporosis, bone damage and repair. METHODS Focused on ADSCs osteogenic differentiation, we conducted mRNA, microRNA expression and bioinformatics analysis, including gene differential expression, time series-based trend analysis, functional enrichment, and generates potential nuclear activating miRNAs (NamiRNA) regulatory network. The screened mRNAs in NamiRNA regulatory network were validated with correlation analysis. RESULTS The NamiRNA Regulatory Network reveals 4 mRNAs (C12orf61, MIR31HG, NFE2L1, and PCYOX1L) significantly downregulated in differentiated group and may be associated with ADSCs stemness. Furthermore, the significantly upregulated 10 genes (ACTA2, TAGLN, LY6E, IFITM3, NGFRAP1, TCEAL4, ATP5C1, CAV1, RPSA, and KDELR3) were significantly enriched in osteogenic-related pathways, and negatively correlated with ADSCs cell stemness in vitro. CONCLUSION These findings uncover potential genes related to ADSCs osteogenic differentiation, and provide theoretical basis for underlying ADSCs osteogenic differentiation and related diseases.
Collapse
Affiliation(s)
- Xin Jin
- Department of Plastic Surgery, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yi Lu
- Department of Plastic Surgery, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Zhihong Fan
- Department of Plastic Surgery, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| |
Collapse
|
2
|
Gou Y, Li A, Dong X, Hao A, Li J, Xiang H, Rahaman S, He TC, Fan J. Lactate transporter MCT4 regulates the hub genes for lipid metabolism and inflammation to attenuate intracellular lipid accumulation in non-alcoholic fatty liver disease. Genes Dis 2025; 12:101554. [PMID: 40330148 PMCID: PMC12052676 DOI: 10.1016/j.gendis.2025.101554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 11/25/2024] [Accepted: 11/25/2024] [Indexed: 05/08/2025] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) patients have multiple metabolic disturbances, with markedly elevated levels of lactate. Lactate accumulations play pleiotropic roles in disease progression through metabolic rearrangements and epigenetic modifications. Monocarboxylate transporter 4 (MCT4) is highly expressed in hepatocytes and responsible for transporting intracellular lactate out of the cell. To explore whether elevated MCT4 levels played any role in NAFLD development, we overexpressed and silenced MCT4 in hepatocytes and performed a comprehensive in vitro and in vivo analysis. Our results revealed that MCT4 overexpression down-regulated the genes for lipid synthesis while up-regulating the genes involved in lipid catabolism. Conversely, silencing MCT4 expression or inhibiting MCT4 expression led to the accumulation of intracellular lipid and glucose metabolites, resulting in hepatic steatosis. In a mouse model of NAFLD, we found that exogenous MCT4 overexpression significantly reduced lipid metabolism and alleviated hepatocellular steatosis. Mechanistically, MCT4 alleviated hepatic steatosis by regulating a group of hub genes such as Arg2, Olr1, Cd74, Mmp8, Irf7, Spp1, and Apoe, which in turn impacted multiple pathways involved in lipid metabolism and inflammatory response, such as PPAR, HIF-1, TNF, IL-17, PI3K-AKT, Wnt, and JAK-STAT. Collectively, our results strongly suggest that MCT4 may play an important role in regulating lipid metabolism and inflammation and thus serve as a potential therapeutic target for NAFLD.
Collapse
Affiliation(s)
- Yannian Gou
- Ministry of Education Key Laboratory of Diagnostic Medicine, and Department of Clinical Biochemistry, School of Clinical Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
- Western Institute of Digital-Intelligent Medicine, Chongqing 401329, China
| | - Aohua Li
- Ministry of Education Key Laboratory of Diagnostic Medicine, and Department of Clinical Biochemistry, School of Clinical Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Xiangyu Dong
- Ministry of Education Key Laboratory of Diagnostic Medicine, and Department of Clinical Biochemistry, School of Clinical Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Ailing Hao
- Ministry of Education Key Laboratory of Diagnostic Medicine, and Department of Clinical Biochemistry, School of Clinical Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Jiajia Li
- Ministry of Education Key Laboratory of Diagnostic Medicine, and Department of Clinical Biochemistry, School of Clinical Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Han Xiang
- Ministry of Education Key Laboratory of Diagnostic Medicine, and Department of Clinical Biochemistry, School of Clinical Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Saidur Rahaman
- Ministry of Education Key Laboratory of Diagnostic Medicine, and Department of Clinical Biochemistry, School of Clinical Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Tong-Chuan He
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Jiaming Fan
- Ministry of Education Key Laboratory of Diagnostic Medicine, and Department of Clinical Biochemistry, School of Clinical Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
- Western Institute of Digital-Intelligent Medicine, Chongqing 401329, China
| |
Collapse
|
3
|
Wu J, Ge Y, Huang W, Zhang L, Huang J, Huang N, Luo Y. Natural bioactive compounds modified with mesenchymal stem cells: new hope for regenerative medicine. Front Bioeng Biotechnol 2025; 13:1446537. [PMID: 40416310 PMCID: PMC12098461 DOI: 10.3389/fbioe.2025.1446537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 04/25/2025] [Indexed: 05/27/2025] Open
Abstract
Mesenchymal stem cells (MSCs) have the potential to differentiate into various cell types, providing important sources of cells for the development of regenerative medicine. Although MSCs have various advantages, there are also various problems, such as the low survival rate of transplanted cells and poor migration and homing; therefore, determining how to reform MSCs to improve their utilization is particularly important. Although many natural bioactive compounds have shown great potential for improving MSCs, many mechanisms and pathways are involved; however, in the final analysis, natural bioactive compounds promoted MSC proliferation, migration and homing and promoted differentiation and antiaging. This article reviews the regulatory effects of natural bioactive compounds on MSCs to provide new ideas for the therapeutic effects of modified MSCs on diseases.
Collapse
Affiliation(s)
- Jingjing Wu
- Department of Neurology, Third Affiliated Hospital of Zunyi Medical University (The First People’s Hospital of Zunyi), Zunyi, Guizhou, China
| | - Ying Ge
- Department of Neurology, Third Affiliated Hospital of Zunyi Medical University (The First People’s Hospital of Zunyi), Zunyi, Guizhou, China
| | - Wendi Huang
- Department of Neurology, Third Affiliated Hospital of Zunyi Medical University (The First People’s Hospital of Zunyi), Zunyi, Guizhou, China
| | - Li Zhang
- Department of Neurology, Third Affiliated Hospital of Zunyi Medical University (The First People’s Hospital of Zunyi), Zunyi, Guizhou, China
| | - Juan Huang
- Key Laboratory of Basic Pharmacology and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou, China
| | - Nanqu Huang
- National Drug Clinical Trial Institution, Third Affiliated Hospital of Zunyi Medical University (The First People’s Hospital of Zunyi), Zunyi, Guizhou, China
- Department of Gerontology, Third Affiliated Hospital of Zunyi Medical University (The First People’s Hospital of Zunyi), Zunyi, Guizhou, China
| | - Yong Luo
- Department of Neurology, Third Affiliated Hospital of Zunyi Medical University (The First People’s Hospital of Zunyi), Zunyi, Guizhou, China
- Department of Gerontology, Third Affiliated Hospital of Zunyi Medical University (The First People’s Hospital of Zunyi), Zunyi, Guizhou, China
| |
Collapse
|
4
|
Wang L, Liu Y, Zhang Y, Sun Q, Wang X, Li Q, Li X. Dual-crosslinkable alginate hydrogel with dynamic viscoelasticity for chondrogenic and osteogenic differentiation of mesenchymal stem cells. Int J Biol Macromol 2025; 307:142346. [PMID: 40118412 DOI: 10.1016/j.ijbiomac.2025.142346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 03/08/2025] [Accepted: 03/19/2025] [Indexed: 03/23/2025]
Abstract
Tissue engineering presents an advanced approach for cartilage and bone tissue repair, with cells serving as a crucial component of the treatment process. The viscoelasticity, a defining fundamental mechanical property, significantly influences cellular behavior. The majority of current research has primarily focused on comparing static elastic and viscoelastic hydrogels with varying stress relaxation rates, while neglecting the inherent dynamic viscoelastic properties of native tissues. Herein, we developed a dynamic viscoelastic hydrogel system employing modified sodium alginate hydrogels to explore the impact of the transfer of viscoelasticity and elastic mechanical properties on the behavior and fate of mesenchymal stem cells (MSCs). The results demonstrated that a viscoelastic environment facilitates greater cell proliferation and spreading. Moreover, extended exposure to the viscoelastic environment resulted in significantly enhanced secretion of osteogenic/chondrogenic extracellular matrix (ECM), upregulates differentiation-specific gene expression, and supports nuclear localization of Yes-associated protein (YAP) and transcriptional co-activator with PDZ-binding motif (TAZ). This study elucidates the mechanical microenvironment required for MSC differentiation, enriching the theoretical foundation for the design of optimized scaffold in cartilage and bone tissue engineering applications.
Collapse
Affiliation(s)
- Litao Wang
- School of Mechanics and Safety Engineering, National Center for International Joint Research of Micro-nano Molding Technology, Zhengzhou University, Zhengzhou 450001, China
| | - Yajing Liu
- School of Mechanics and Safety Engineering, National Center for International Joint Research of Micro-nano Molding Technology, Zhengzhou University, Zhengzhou 450001, China
| | - Yuhang Zhang
- School of Mechanics and Safety Engineering, National Center for International Joint Research of Micro-nano Molding Technology, Zhengzhou University, Zhengzhou 450001, China
| | - Qingqing Sun
- School of Materials Science and Engineering, Zhengzhou Key Laboratory of Flexible Electronic Materials and Thin-Film Technologies, Zhengzhou University, Zhengzhou 450001, China
| | - Xiaofeng Wang
- School of Mechanics and Safety Engineering, National Center for International Joint Research of Micro-nano Molding Technology, Zhengzhou University, Zhengzhou 450001, China
| | - Qian Li
- School of Mechanics and Safety Engineering, National Center for International Joint Research of Micro-nano Molding Technology, Zhengzhou University, Zhengzhou 450001, China.
| | - Xiaomeng Li
- School of Mechanics and Safety Engineering, National Center for International Joint Research of Micro-nano Molding Technology, Zhengzhou University, Zhengzhou 450001, China.
| |
Collapse
|
5
|
Dong X, Xiang H, Li J, Hao A, Wang H, Gou Y, Li A, Rahaman S, Qiu Y, Li J, Mei O, Zhong J, You W, Shen G, Wu X, Li J, Shu Y, Shi LL, Zhu Y, Reid RR, He TC, Fan J. Dermal fibroblast-derived extracellular matrix (ECM) synergizes with keratinocytes in promoting re-epithelization and scarless healing of skin wounds: Towards optimized skin tissue engineering. Bioact Mater 2025; 47:1-17. [PMID: 39872210 PMCID: PMC11762682 DOI: 10.1016/j.bioactmat.2024.12.030] [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: 09/20/2024] [Revised: 12/24/2024] [Accepted: 12/27/2024] [Indexed: 01/30/2025] Open
Abstract
Skin serves as the first-order protective barrier against the environment and any significant disruptions in skin integrity must be promptly restored. Despite significant advances in therapeutic strategies, effective management of large chronic skin wounds remains a clinical challenge. Dermal fibroblasts are the primary cell type responsible for remodeling the extracellular matrix (ECM) in wound healing. Here, we investigated whether ECM derived from exogenous fibroblasts, in combination with keratinocytes, promoted scarless cutaneous wound healing. To overcome the limited lifespan of primary dermal fibroblasts, we established reversibly immortalized mouse dermal fibroblasts (imDFs), which were non-tumorigenic, expressed dermal fibroblast markers, and were responsive to TGF-β1 stimulation. The decellularized ECM prepared from both imDFs and primary dermal fibroblasts shared similar expression profiles of extracellular matrix proteins and promoted the proliferation of keratinocyte (iKera) cells. The imDFs-derived ECM solicited no local immune response. While the ECM and to a lesser extent imDFs enhanced skin wound healing with excessive fibrosis, a combination of imDFs-derived ECM and iKera cells effectively promoted the re-epithelization and scarless healing of full-thickness skin wounds. These findings strongly suggest that dermal fibroblast-derived ECM, not fibroblasts themselves, may synergize with keratinocytes in regulating scarless healing and re-epithelialization of skin wounds. Given its low immunogenic nature, imDFs-derived ECM should be a valuable resource of skin-specific biomaterial for wound healing and skin tissue engineering.
Collapse
Affiliation(s)
- Xiangyu Dong
- Ministry of Education Key Laboratory of Diagnostic Medicine, and Department of Clinical Biochemistry, School of Clinical Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Han Xiang
- Ministry of Education Key Laboratory of Diagnostic Medicine, and Department of Clinical Biochemistry, School of Clinical Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Jiajia Li
- Ministry of Education Key Laboratory of Diagnostic Medicine, and Department of Clinical Biochemistry, School of Clinical Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Ailing Hao
- Ministry of Education Key Laboratory of Diagnostic Medicine, and Department of Clinical Biochemistry, School of Clinical Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Hao Wang
- Ministry of Education Key Laboratory of Diagnostic Medicine, and Department of Clinical Biochemistry, School of Clinical Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA
| | - Yannian Gou
- Ministry of Education Key Laboratory of Diagnostic Medicine, and Department of Clinical Biochemistry, School of Clinical Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Aohua Li
- Ministry of Education Key Laboratory of Diagnostic Medicine, and Department of Clinical Biochemistry, School of Clinical Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Saidur Rahaman
- Ministry of Education Key Laboratory of Diagnostic Medicine, and Department of Clinical Biochemistry, School of Clinical Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Yiheng Qiu
- Ministry of Education Key Laboratory of Diagnostic Medicine, and Department of Clinical Biochemistry, School of Clinical Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Jiahao Li
- Ministry of Education Key Laboratory of Diagnostic Medicine, and Department of Clinical Biochemistry, School of Clinical Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Ou Mei
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA
- Department of Orthopedic Surgery, Jiangxi Hospital of Traditional Chinese Medicine, Jiangxi University of Traditional Chinese Medicine, Nanchang, 330006, China
| | - Jiamin Zhong
- Ministry of Education Key Laboratory of Diagnostic Medicine, and Department of Clinical Biochemistry, School of Clinical Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA
| | - Wulin You
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA
- Department of Orthopaedic Surgery, Wuxi Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi, 214071, China
| | - Guowei Shen
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA
- Department of Orthopaedic Surgery, BenQ Medical Center, The Affiliated BenQ Hospital of Nanjing Medical University, Nanjing, 210019, China
| | - Xingye Wu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA
- Department of Gastrointestinal Surgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Jingjing Li
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA
- Department of Oncology, The Affiliated Hospital of Shandong Second Medical University, Weifang, 261053, China
| | - Yi Shu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA
- Stem Cell Biology and Therapy Laboratory of the Pediatric Research Institute, the National Clinical Research Center for Child Health and Disorders, and Ministry of Education Key Laboratory of Child Development and Disorders, the Children's Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Lewis L. Shi
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA
| | - Yi Zhu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA
| | - Russell R. Reid
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, 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
| | - Tong-Chuan He
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, 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
| | - Jiaming Fan
- Ministry of Education Key Laboratory of Diagnostic Medicine, and Department of Clinical Biochemistry, School of Clinical Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
- Western Institute of Digital-Intelligent Medicine, Chongqing, 401329, China
| |
Collapse
|
6
|
Wang B, Lyu FJ, Deng Z, Zheng Q, Ma Y, Peng Y, Guo S, Lei G, Lai Y, Li Q. Therapeutic potential of stem cell-derived exosomes for bone tissue regeneration around prostheses. J Orthop Translat 2025; 52:85-96. [PMID: 40291635 PMCID: PMC12023751 DOI: 10.1016/j.jot.2025.03.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 03/09/2025] [Accepted: 03/14/2025] [Indexed: 04/30/2025] Open
Abstract
Artificial joint replacement is a widely recognized treatment for arthritis and other severe joint conditions. However, one of the primary causes of failure in joint replacements is the loosening of the prosthesis. After implantation, wear particles between the implant and the adjacent bone tissue are the principal contributors to this loosening. Recently, exosomes have garnered significant interest due to their low immunogenicity and effective membrane binding. They have shown potential in promoting bone regeneration via the paracrine pathway. This review examines the role and mechanisms of exosomes derived from mesenchymal stem cells (MSCs) in bone regeneration, their impact on the integration of various implants into surrounding bone tissue and current challenges and future directions for the clinical application of exosomes. The Translational Potential of this Article: Emerging evidence suggests that mesenchymal stem cell-derived exosomes may offer a promising therapeutic strategy for aseptic prosthesis loosening, potentially mediated through mechanisms such as modulation of inflammatory responses, suppression of osteoclastogenesis, enhancement of osteogenic differentiation and facilitation of bone regeneration. Preclinical studies further indicate that the therapeutic potential of these extracellular vesicles could be optimized through bioengineering strategies, including surface modification and cargo-loading techniques, warranting further investigation to advance their clinical translation.
Collapse
Affiliation(s)
- Biwu Wang
- Department of Orthopedics, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, China
| | - Feng-Juan Lyu
- The Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, 510000, China
- South China University of Technology-The University of Western Australia Joint Center for Regenerative Medicine Research, School of Medicine, South China University of Technology, Guangzhou, 510006, China
| | - Zhantao Deng
- Department of Orthopedics, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Qiujian Zheng
- Department of Orthopedics, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Yuanchen Ma
- Department of Orthopedics, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Yujie Peng
- Department of Orthopedics, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
- Shantou University Medical College, Xinling Road 22, Shantou, 515041, China
| | - Shujun Guo
- South China University of Technology-The University of Western Australia Joint Center for Regenerative Medicine Research, School of Medicine, South China University of Technology, Guangzhou, 510006, China
| | - Guihua Lei
- South China University of Technology-The University of Western Australia Joint Center for Regenerative Medicine Research, School of Medicine, South China University of Technology, Guangzhou, 510006, China
| | - Yonggang Lai
- South China University of Technology-The University of Western Australia Joint Center for Regenerative Medicine Research, School of Medicine, South China University of Technology, Guangzhou, 510006, China
| | - Qingtian Li
- Department of Orthopedics, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| |
Collapse
|
7
|
Yang H, Yang H, Wang Q, Ji H, Qian T, Qiao Y, Shi J, Cong M. Mesenchymal stem cells and their extracellular vesicles: new therapies for cartilage repair. Front Bioeng Biotechnol 2025; 13:1591400. [PMID: 40343207 PMCID: PMC12058886 DOI: 10.3389/fbioe.2025.1591400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2025] [Accepted: 04/08/2025] [Indexed: 05/11/2025] Open
Abstract
Cartilage is crucial for joints, and its damage can lead to pain and functional impairment, causing financial burden to patients. Due to its weak self-repair, cartilage injury control is a research focus. Cartilage injury naturally with age, but mechanical trauma, lifestyle factors and certain genetic abnormalities can increase the likelihood of symptomatic disease progression. Current treatments for cartilage injury include pharmacological and surgical interventions, but these lack the ability to stop the progression of disease and restore the regeneration of the cartilage. Biological therapies have been evaluated but show varying degrees of efficacy in cartilage regeneration long-term. The mesenchymal stem cell (MSC) therapy attracts attention as it is easily harvested and expanded. Once thought to repair via differentiation, MSCs are now known to secrete extracellular vesicles (EVs) paracrinely. These EVs, rich in bioactive molecules, enable cell communication, boost growth factor secretion, regulate the synthesis and degradation of extracellular matrix (ECM), and modulate inflammation, vital for cartilage repair. However, further research and clinical validation are still required for the application of MSC and MSC-EVs. This review highlights the current state of research on the use of MSC and MSC-EVs in the treatment of cartilage injury. It is hoped that the review in this paper will provide valuable references and inspiration for future researchers in therapeutic studies of cartilage repair.
Collapse
Affiliation(s)
- Hongwei Yang
- Department of Orthopedics, Affiliated Nantong Hospital 3 of Nantong University, Nantong, China
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education and Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Haochen Yang
- School of Medicine, Nantong University, Nantong, China
| | - Qin Wang
- Department of Orthopedics, Affiliated Nantong Hospital 3 of Nantong University, Nantong, China
| | - Hanzhen Ji
- Department of Orthopedics, Affiliated Nantong Hospital 3 of Nantong University, Nantong, China
| | - Tianmei Qian
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education and Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, China
- Engineering Research Center of Integration and Application of Digital Learning Technology, Ministry of Education, Beijing, China
| | - Yusen Qiao
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Junfeng Shi
- Department of Orthopedics, Affiliated Nantong Hospital 3 of Nantong University, Nantong, China
| | - Meng Cong
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education and Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, China
| |
Collapse
|
8
|
Shao C, Chen H, Liu T, Pan C. The Hippo pathway in bone and cartilage: implications for development and disease. PeerJ 2025; 13:e19334. [PMID: 40292098 PMCID: PMC12024444 DOI: 10.7717/peerj.19334] [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: 11/13/2024] [Accepted: 03/26/2025] [Indexed: 04/30/2025] Open
Abstract
Bone is the main structure of the human body; it mainly plays a supporting role and participates in metabolic processes. The Hippo signaling pathway is composed of a series of protein kinases, including the mammalian STE20-like kinase MST1/2 and the large tumor suppressor LATS1/2, which are widely involved in pathophysiological processes, including cell proliferation, differentiation, apoptosis and death, especially those related to biomechanical transduction in vivo. However, the role of it in regulating skeletal system development and the evolution of bone-related diseases remains poorly understood. The pathway can intervene in and regulate the physiological activities of bone-related cells such as osteoclasts and chondrocytes through its own or other bone-related signaling pathways, such as the Wnt pathway, the Notch pathway, and receptor activator of nuclear factor-κB ligand (RANKL), thereby affecting the occurrence and development of bone diseases. This article discusses the role of the Hippo signaling pathway in bone development and disease to provide new insights into the treatment of bone-related diseases by targeting the Hippo signaling pathway.
Collapse
Affiliation(s)
- Chenwei Shao
- Institute of Translational Medicine, Yangzhou University, Yangzhou, Jiangsu, China
| | - Hao Chen
- Institute of Translational Medicine, Yangzhou University, Yangzhou, Jiangsu, China
- Department of Orthopedics, Affiliated Hospital of Yangzhou University, Yangzhou, Jiangsu, China
| | - Tingting Liu
- Department of Orthopedics, Affiliated Hospital of Yangzhou University, Yangzhou, Jiangsu, China
| | - Chun Pan
- Institute of Translational Medicine, Yangzhou University, Yangzhou, Jiangsu, China
- Department of Orthopedics, Affiliated Hospital of Yangzhou University, Yangzhou, Jiangsu, China
| |
Collapse
|
9
|
Dalle Carbonare L, Cominacini M, Trabetti E, Bombieri C, Pessoa J, Romanelli MG, Valenti MT. The bone microenvironment: new insights into the role of stem cells and cell communication in bone regeneration. Stem Cell Res Ther 2025; 16:169. [PMID: 40221779 PMCID: PMC11993959 DOI: 10.1186/s13287-025-04288-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Accepted: 03/19/2025] [Indexed: 04/14/2025] Open
Abstract
Mesenchymal stem cells (MSCs) play a crucial role in bone formation and remodeling. Intrinsic genetic factors and extrinsic environmental cues regulate their differentiation into osteoblasts. Within the bone microenvironment, a complex network of biochemical and biomechanical signals orchestrates bone homeostasis and regeneration. In addition, the crosstalk among MSCs, immune cells, and neighboring cells-mediated by extracellular vesicles and non-coding RNAs (such as circular RNAs and micro RNAs) -profoundly influences osteogenic differentiation and bone remodeling. Recent studies have explored specific signaling pathways that contribute to effective bone regeneration, highlighting the potential of manipulating the bone microenvironment to enhance MSC functionality. The integration of advanced biomaterials, gene editing techniques, and controlled delivery systems is paving the way for more targeted and efficient regenerative therapies. Furthermore, artificial intelligence could improve bone tissue engineering, optimize biomaterial design, and enable personalized treatment strategies. This review explores the latest advancements in bone regeneration, emphasizing the intricate interplay among stem cells, immune cells, and signaling molecules. By providing a comprehensive overview of these mechanisms and their clinical implications, we aim to shed light on future research directions in this rapidly evolving field.
Collapse
Affiliation(s)
- L Dalle Carbonare
- Department of Engineering for the Innovation Medicine, University of Verona, 37100, Verona, Italy
| | - M Cominacini
- Department of Engineering for the Innovation Medicine, University of Verona, 37100, Verona, Italy
| | - E Trabetti
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, 37100, Verona, Italy
| | - C Bombieri
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, 37100, Verona, Italy
| | - J Pessoa
- Department of Medical Sciences and Institute of Biomedicine-Ibimed, University of Aveiro, 3810 - 193, Aveiro, Portugal
| | - M G Romanelli
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, 37100, Verona, Italy
| | - M T Valenti
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, 37100, Verona, Italy.
| |
Collapse
|
10
|
Zhu Y, Liu Q, Yu C, Zhang H, Zhong J, Wang Y, Mei O, Gerhard E, You W, Shen G, Luo C, Wu X, Li J, Shu Y, Wen Y, Zeb U, Luu HH, Lee MJ, Shi LL, Bi Y, Yang J, Fan J, Reid RR, He T, Wen L. An Intervertebral Disc (IVD) Regeneration Model Using Human Nucleus Pulposus Cells (iHNPCs) and Annulus Fibrosus Cells (iHAFCs). Adv Healthc Mater 2025; 14:e2403742. [PMID: 40052622 PMCID: PMC12004445 DOI: 10.1002/adhm.202403742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 01/26/2025] [Indexed: 04/18/2025]
Abstract
Intervertebral disc (IVD) degeneration (IVDD), primarily caused by nucleus pulposus (NP) dehydration, leads to low back pain. While current treatments focus on symptom management or surgical intervention, tissue engineering using IVD-derived cells, biofactors, and scaffolds offers a promising regenerative approach. Here, human NP cells (NPCs) and annulus fibrosus cells (AFCs) are immortalized with human telomerase reverse transcriptase (hTERT), generating immortalized NPCs (iHNPCs) and AFCs (iHAFCs). These cells express NP and AF-specific markers, are reversible via FLP recombinase, and are non-tumorigenic. iHAFCs exhibit osteogenic potential, while iHNPCs show chondrogenic differentiation. A 3D-printed citrate-based scaffold was employed to develop an IVD regeneration model, with BMP9-stimulated iHAFCs in the peripheral region and BMP2-stimulated iHNPCs in the central region. Histological analysis revealed bone formation in the iHAFC region and cartilage formation in the iHNPC region, mimicking the natural IVD structure. Additionally, an ex vivo spine fusion model demonstrated robust bone formation in iHAFC-treated segments. These findings highlight the potential of iHAFCs and iHNPCs as valuable tools for IVD tissue engineering and regeneration.
Collapse
|
11
|
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
|
12
|
Zhang L, Yu Z, Liu S, Liu F, Zhou S, Zhang Y, Tian Y. Advanced progress of adipose-derived stem cells-related biomaterials in maxillofacial regeneration. Stem Cell Res Ther 2025; 16:110. [PMID: 40038758 PMCID: PMC11881347 DOI: 10.1186/s13287-025-04191-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Accepted: 01/24/2025] [Indexed: 03/06/2025] Open
Abstract
The tissue injury in maxillofacial region affects patients' physical function and specific mental health. This decade, utilizing regenerative medicine to achieve tissue regeneration has been proved a hopeful direction. Seed cells play a vital role in regeneration strategy. Among various kinds of stem cells that effectively to regenerate the soft and hard tissue of maxillofacial region, adipose-derived stem cells (ADSCs) have gained increasing interests of researchers due to their abundant sources, easy availability and multi-differentiation potentials in recent decades. Thus, this review focuses on the advances of ADSCs-based biomaterial in maxillofacial regeneration from the progress and strategies perspective. It is structured as introducing the properties of ADSCs, biomaterials (polymers, ceramics and metals) within ADSCs and the latest applications of ADSCs in maxillofacial regeneration, including temporomandibular joint (TMJ), bone, periodontal tissue, tooth, nerve as well as cosmetic field. In order to further facilitate ADSCs-based therapies as an emerging platform for regenerative medicine, this review also emphasized current challenges in translating ADSC-based therapies into clinical application and dissussed the strategies to solve these obstacles.
Collapse
Affiliation(s)
- Lijun Zhang
- Department of Orthodontics, School and Hospital of Stomatology, China Medical University, Nanjing North Street 117, Shenyang, 110002, China
| | - Zihang Yu
- Department of Orthodontics, School and Hospital of Stomatology, China Medical University, Nanjing North Street 117, Shenyang, 110002, China
| | - Shuchang Liu
- Department of Orthodontics, School and Hospital of Stomatology, China Medical University, Nanjing North Street 117, Shenyang, 110002, China
| | - Fan Liu
- Department of Orthodontics, School and Hospital of Stomatology, China Medical University, Nanjing North Street 117, Shenyang, 110002, China
| | - Shijie Zhou
- Department of Orthodontics, School and Hospital of Stomatology, China Medical University, Nanjing North Street 117, Shenyang, 110002, China
| | - Yuanyuan Zhang
- Department of Orthodontics, School and Hospital of Stomatology, China Medical University, Nanjing North Street 117, Shenyang, 110002, China
| | - Yulou Tian
- Department of Orthodontics, School and Hospital of Stomatology, China Medical University, Nanjing North Street 117, Shenyang, 110002, China.
| |
Collapse
|
13
|
Wang L, Ruan M, Bu Q, Zhao C. Signaling Pathways Driving MSC Osteogenesis: Mechanisms, Regulation, and Translational Applications. Int J Mol Sci 2025; 26:1311. [PMID: 39941080 PMCID: PMC11818554 DOI: 10.3390/ijms26031311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 01/23/2025] [Accepted: 01/25/2025] [Indexed: 02/16/2025] Open
Abstract
Mesenchymal stem cells (MSCs) are crucial for skeletal development, homeostasis, and repair, primarily through their differentiation into osteoblasts and other skeletal lineage cells. Key signaling pathways, including Wnt, TGF-β/BMP, PTH, Hedgehog, and IGF, act as critical regulators of MSC osteogenesis, playing pivotal roles in maintaining bone homeostasis and facilitating regeneration. These pathways interact in distinct ways at various stages of bone development, mineralization, and remodeling. This review provides an overview of the molecular mechanisms by which these pathways regulate MSC osteogenesis, their influence on bone tissue formation, and their implications in bone diseases and therapeutic strategies. Additionally, we explore the potential applications of these pathways in bone tissue engineering, with a particular focus on promoting the use of MSCs as seed cells for bone defect repair. Ultimately, this review aims to highlight potential avenues for advancing bone biology research, treating bone disorders, and enhancing regenerative medicine.
Collapse
Affiliation(s)
| | | | | | - Chengzhu Zhao
- Laboratory of Skeletal Development and Regeneration, Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| |
Collapse
|
14
|
Veret D, Tejedor G, Perez E, Chomette A, Farno M, Ferreira-Lopez R, Dagneaux L, Pers YM, Jorgsensen C, Gondeau C, Brondello JM. Combination of rapamycin and adipose-derived mesenchymal stromal cells enhances therapeutic potential for osteoarthritis. Stem Cell Res Ther 2025; 16:9. [PMID: 39815291 PMCID: PMC11737215 DOI: 10.1186/s13287-024-04090-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 12/03/2024] [Indexed: 01/30/2025] Open
Abstract
BACKGROUND The regenerative potential of mesenchymal stromal/stem cells (MSCs) has been extensively studied in clinical trials in the past decade. However, despite the promising regenerative properties documented in preclinical studies, for instance in osteoarthritis (OA), the therapeutic translation of these results in patients has not been fully conclusive. One factor contributing to this therapeutic barrier could be the presence of senescent cells in OA joints. METHODS This study evaluated a novel approach to OA treatment by combining adipose tissue-derived MSCs (AD-MSCs) with rapamycin, a clinically approved immunosuppressive drug with anti-senescence properties. First, rapamycin effects on senescence and fibrosis markers were investigated in freshly isolated OA chondrocytes by immunostaining. Next, the in vitro differentiation capacities of AD-MSCs, their regulatory immune functions on activated immune cells and their regenerative effects on OA chondrocyte signature were assessed in the presence of rapamycin. RESULTS In OA chondrocytes, rapamycin reduced the senescence marker p15INK4B and the fibrosis marker COL1A1 without affecting the expression of the master chondrogenic markers SOX9 and COL2. Rapamycin also enhanced AD-MSC differentiation into chondrocytes and reduced their differentiation into adipocytes. In addition, rapamycin improved AD-MSC immunoregulatory functions by promoting the expression of immunosuppressive factors, such as IDO1, PTGS2 and also CD274 (encoding PD-L1). Finally, RNA sequencing analysis showed that in the presence of rapamycin, AD-MSCs displayed improved chondroprotective regenerative effects on co-cultured OA chondrocytes. CONCLUSIONS Our findings suggest that the rapamycin and AD-MSC combination enhances the therapeutic efficacy of these cells in senescence-driven degenerative diseases such as OA, notably by improving their anti-fibrotic and anti-inflammatory properties.
Collapse
Affiliation(s)
- Damien Veret
- IRMB, Univ Montpellier, INSERM, CHU St Eloi, 80 AV A Fliche, 34295-Cedex-05, Montpellier, France
- MedXCell, IRMB, CHU St Eloi, Cyborg, Montpellier, France
| | | | - Esther Perez
- MedXCell, IRMB, CHU St Eloi, Cyborg, Montpellier, France
| | | | - Maylis Farno
- MedXCell, IRMB, CHU St Eloi, Cyborg, Montpellier, France
| | - Rosanna Ferreira-Lopez
- IRMB, Univ Montpellier, INSERM, CHU St Eloi, 80 AV A Fliche, 34295-Cedex-05, Montpellier, France
- Clinical Immunology and Osteoarticular Diseases Therapeutic Unit, Hôpital Lapeyronie, Montpellier, France
- Rheumatology department, Regional Narbonne Hospital, Narbonne, France
| | - Louis Dagneaux
- Hôpital Lapeyronie, Orthopedic Service, Montpellier, France
| | - Yves-Marie Pers
- IRMB, Univ Montpellier, INSERM, CHU St Eloi, 80 AV A Fliche, 34295-Cedex-05, Montpellier, France
- Clinical Immunology and Osteoarticular Diseases Therapeutic Unit, Hôpital Lapeyronie, Montpellier, France
| | - Christian Jorgsensen
- IRMB, Univ Montpellier, INSERM, CHU St Eloi, 80 AV A Fliche, 34295-Cedex-05, Montpellier, France
- Clinical Immunology and Osteoarticular Diseases Therapeutic Unit, Hôpital Lapeyronie, Montpellier, France
| | - Claire Gondeau
- MedXCell, IRMB, CHU St Eloi, Cyborg, Montpellier, France
| | - Jean-Marc Brondello
- IRMB, Univ Montpellier, INSERM, CHU St Eloi, 80 AV A Fliche, 34295-Cedex-05, Montpellier, France.
| |
Collapse
|
15
|
Zhao P, Zhu Y, Kim M, Zhao G, Wang Y, Collins CP, Mei O, Zhang Y, Duan C, Zhong J, Zhang H, You W, Shen G, Luo C, Wu X, Li J, Shu Y, Luu HH, Haydon RC, Lee MJ, Shi LL, Huang W, Fan J, Sun C, Wen L, Ameer GA, He TC, Reid RR. Effective Bone Tissue Fabrication Using 3D-Printed Citrate-Based Nanocomposite Scaffolds Laden with BMP9-Stimulated Human Urine Stem Cells. ACS APPLIED MATERIALS & INTERFACES 2025; 17:197-210. [PMID: 39718997 PMCID: PMC11783527 DOI: 10.1021/acsami.4c13246] [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: 08/06/2024] [Revised: 10/30/2024] [Accepted: 10/30/2024] [Indexed: 12/26/2024]
Abstract
Effective repair of large bone defects through bone tissue engineering (BTE) remains an unmet clinical challenge. Successful BTE requires optimal and synergistic interactions among biocompatible scaffolds, osteogenic factors, and osteoprogenitors to form a highly vascularized microenvironment for bone regeneration and osseointegration. We sought to develop a highly effective BTE system by using 3D printed citrate-based mPOC/hydroxyapatite (HA) composites laden with BMP9-stimulated human urine stem cells (USCs). Specifically, we synthesized and characterized methacrylate poly(1,8 octamethylene citrate) (mPOC), mixed it with 0%, 40% or 60% HA (i.e., mPOC-0HA, mPOC-40HA, or mPOC-60HA), and fabricated composite scaffold via micro-continuous liquid interface production (μCLIP). The 3D-printed mPOC-HA composite scaffolds were compatible with human USCs that exhibited high osteogenic activity in vitro upon BMP9 stimulation. Subcutaneous implantation of mPOC-HA scaffolds laden with BMP9-stimulated USCs revealed effective bone formation in all three types of mPOC-HA composite scaffolds. Histologic evaluation revealed that the mPOC-60HA composite scaffold yielded the most mature bone, resembling native bone tissue with extensive scaffold-osteointegration. Collectively, these findings demonstrate that the citrate-based mPOC-60HA composite, human urine stem cells, and the potent osteogenic factor BMP9 constitute a desirable triad for effective bone tissue engineering.
Collapse
Affiliation(s)
- Piao Zhao
- Departments
of Orthopaedic Surgery, Urology, and Gastrointestinal Surgery, the First Affiliated Hospital of Chongqing Medical
University, Chongqing 400016, China
- Molecular
Oncology Laboratory, Department of Orthopedic Surgery and Rehabilitation
Medicine, The University of Chicago Medical
Center; Chicago, Illinois 60637, United States
| | - Yi Zhu
- Molecular
Oncology Laboratory, Department of Orthopedic Surgery and Rehabilitation
Medicine, The University of Chicago Medical
Center; Chicago, Illinois 60637, United States
- Department
of Orthopaedic Surgery, Beijing Hospital,
National Center of Gerontology, Chinese Academy of Medical Sciences
& Peking Union Medical College, Beijing, 100005, China
| | - Mirae Kim
- Department
of Biomedical Engineering, Northwestern
University; Evanston, Illinois 60208, United States
- Center
for Advanced Regenerative Engineering, Northwestern
University, Evanston, Illinois 60208, United States
| | - Guozhi Zhao
- Departments
of Orthopaedic Surgery, Urology, and Gastrointestinal Surgery, the First Affiliated Hospital of Chongqing Medical
University, Chongqing 400016, China
- Molecular
Oncology Laboratory, Department of Orthopedic Surgery and Rehabilitation
Medicine, The University of Chicago Medical
Center; Chicago, Illinois 60637, United States
| | - Yonghui Wang
- Molecular
Oncology Laboratory, Department of Orthopedic Surgery and Rehabilitation
Medicine, The University of Chicago Medical
Center; Chicago, Illinois 60637, United States
- Department
of Geriatrics, Xinhua Hospital, Shanghai
Jiao-Tong University School of Medicine, Shanghai 200000, China
| | - Caralyn P. Collins
- Department
of Mechanical Engineering, Northwestern
University; Evanston, Illinois 60208, United States
| | - Ou Mei
- Molecular
Oncology Laboratory, Department of Orthopedic Surgery and Rehabilitation
Medicine, The University of Chicago Medical
Center; Chicago, Illinois 60637, United States
- Department
of Orthopedic Surgery, Jiangxi Hospital of Traditional Chinese Medicine, Jiangxi University of Traditional Chinese Medicine, Nanchang 330006, China
| | - Yuanyuan Zhang
- Wake Forest
Institute for Regenerative Medicine, Wake
Forest University School of Medicine, Winston-Salem, North Carolina 27101, United States
| | - Chongwen Duan
- Department
of Biomedical Engineering, Northwestern
University; Evanston, Illinois 60208, United States
| | - Jiamin Zhong
- Molecular
Oncology Laboratory, Department of Orthopedic Surgery and Rehabilitation
Medicine, The University of Chicago Medical
Center; Chicago, Illinois 60637, United States
- Ministry
of Education Key Laboratory of Diagnostic Medicine, and Department
of Clinical Biochemistry, School of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Hui Zhang
- Molecular
Oncology Laboratory, Department of Orthopedic Surgery and Rehabilitation
Medicine, The University of Chicago Medical
Center; Chicago, Illinois 60637, United States
- The Breast
Cancer Center, Chongqing University Cancer
Hospital, Chongqing 4000430, China
| | - Wulin You
- Molecular
Oncology Laboratory, Department of Orthopedic Surgery and Rehabilitation
Medicine, The University of Chicago Medical
Center; Chicago, Illinois 60637, United States
- Department
of Orthopaedic Surgery, Wuxi Hospital Affiliated
to Nanjing University of Chinese Medicine, Wuxi 214071, China
| | - Guowei Shen
- Molecular
Oncology Laboratory, Department of Orthopedic Surgery and Rehabilitation
Medicine, The University of Chicago Medical
Center; Chicago, Illinois 60637, United States
- Department
of Orthopaedic Surgery, BenQ Medical Center, The Affiliated BenQ Hospital of Nanjing Medical University, Nanjing 210019, China
| | - Changqi Luo
- Molecular
Oncology Laboratory, Department of Orthopedic Surgery and Rehabilitation
Medicine, The University of Chicago Medical
Center; Chicago, Illinois 60637, United States
- Department
of Orthopaedic Surgery, Yibin Second People’s
Hospital, Affiliated with West China School of Medicine, Yibin 644000, China
| | - Xingye Wu
- Departments
of Orthopaedic Surgery, Urology, and Gastrointestinal Surgery, the First Affiliated Hospital of Chongqing Medical
University, Chongqing 400016, China
- Molecular
Oncology Laboratory, Department of Orthopedic Surgery and Rehabilitation
Medicine, The University of Chicago Medical
Center; Chicago, Illinois 60637, United States
| | - Jingjing Li
- Molecular
Oncology Laboratory, Department of Orthopedic Surgery and Rehabilitation
Medicine, The University of Chicago Medical
Center; Chicago, Illinois 60637, United States
- Department
of Oncology, The Affiliated Hospital of
Shandong Second Medical University, Weifang 261053, China
| | - Yi Shu
- Molecular
Oncology Laboratory, Department of Orthopedic Surgery and Rehabilitation
Medicine, The University of Chicago Medical
Center; Chicago, Illinois 60637, United States
- Stem Cell
Biology and Therapy Laboratory of the Pediatric Research Institute,
the National Clinical Research Center for Child Health and Disorders,
and Ministry of Education Key Laboratory of Child Development and
Disorders, the Children’s Hospital
of Chongqing Medical University, Chongqing 400016, China
| | - Hue H. Luu
- Molecular
Oncology Laboratory, Department of Orthopedic Surgery and Rehabilitation
Medicine, The University of Chicago Medical
Center; Chicago, Illinois 60637, United States
| | - Rex C. Haydon
- Molecular
Oncology Laboratory, Department of Orthopedic Surgery and Rehabilitation
Medicine, The University of Chicago Medical
Center; Chicago, Illinois 60637, United States
| | - Michael J. Lee
- Molecular
Oncology Laboratory, Department of Orthopedic Surgery and Rehabilitation
Medicine, The University of Chicago Medical
Center; Chicago, Illinois 60637, United States
| | - Lewis L. Shi
- Molecular
Oncology Laboratory, Department of Orthopedic Surgery and Rehabilitation
Medicine, The University of Chicago Medical
Center; Chicago, Illinois 60637, United States
| | - Wei Huang
- Departments
of Orthopaedic Surgery, Urology, and Gastrointestinal Surgery, the First Affiliated Hospital of Chongqing Medical
University, Chongqing 400016, China
| | - Jiaming Fan
- Molecular
Oncology Laboratory, Department of Orthopedic Surgery and Rehabilitation
Medicine, The University of Chicago Medical
Center; Chicago, Illinois 60637, United States
- Ministry
of Education Key Laboratory of Diagnostic Medicine, and Department
of Clinical Biochemistry, School of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Cheng Sun
- Department
of Mechanical Engineering, Northwestern
University; Evanston, Illinois 60208, United States
- Center
for Advanced Regenerative Engineering, Northwestern
University, Evanston, Illinois 60208, United States
| | - Liangyuan Wen
- Department
of Orthopaedic Surgery, Beijing Hospital,
National Center of Gerontology, Chinese Academy of Medical Sciences
& Peking Union Medical College, Beijing, 100005, China
| | - Guillermo A. Ameer
- Department
of Biomedical Engineering, Northwestern
University; Evanston, Illinois 60208, United States
- Center
for Advanced Regenerative Engineering, Northwestern
University, Evanston, Illinois 60208, United States
- Department
of Surgery, Feinberg School of Medicine, Chicago, Illinois 60611, United States
| | - Tong-Chuan He
- Molecular
Oncology Laboratory, Department of Orthopedic Surgery and Rehabilitation
Medicine, The University of Chicago Medical
Center; Chicago, Illinois 60637, United States
- Center
for Advanced Regenerative Engineering, Northwestern
University, Evanston, Illinois 60208, United States
- Laboratory
of Craniofacial Biology and Development, Section of Plastic and Reconstructive
Surgery, Department of Surgery, The University
of Chicago Medical Center, Chicago, Illinois 60637, United States
| | - Russell R. Reid
- Molecular
Oncology Laboratory, Department of Orthopedic Surgery and Rehabilitation
Medicine, The University of Chicago Medical
Center; Chicago, Illinois 60637, United States
- Center
for Advanced Regenerative Engineering, Northwestern
University, Evanston, Illinois 60208, United States
- Laboratory
of Craniofacial Biology and Development, Section of Plastic and Reconstructive
Surgery, Department of Surgery, The University
of Chicago Medical Center, Chicago, Illinois 60637, United States
| |
Collapse
|
16
|
Zhou H, Ren Y, Zou K, Jin Y, Liu H, Jiang H, Shi L, Sheng X, Weeks J, Wang H, Xue T, Schwarz EM, Xie C, Deng Z, Wang L, Chu L. Efficacy of pH-Responsive Surface Functionalized Titanium Screws in Treating Implant-associated S. aureus Osteomyelitis with Biofilms Formation. Adv Healthc Mater 2025; 14:e2403261. [PMID: 39604325 PMCID: PMC11773098 DOI: 10.1002/adhm.202403261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 11/14/2024] [Indexed: 11/29/2024]
Abstract
Implant-associated Staphylococcus aureus (S. aureus) osteomyelitis (IASO) leads to high orthopedic implant failure rates due to the formation of Staphylococcal abscess community within the bone marrow and bacterial colonization in the osteocyte lacuno-canalicular network (OLCN). To address this, antimicrobial peptides (HHC36)-loaded titania nanotubes (NTs) are developed on titanium screws (Ti-NTs-P-A), which integrate pH-responsive polymethacrylic acid to control HHC36 release for eradicating bacteria in IASO. Colony-forming unit assay confirmed that Ti-NTs-P-A screws maintained sustainable antibacterial effectiveness, killing over 65% of S. aureus even after multiple bacterial solution replacements. Notably, Ti-NTs-P-A screws exhibit significant pH-responsive HHC36 release behavior and bactericidal activity, consistent with the phenotype of peptides-killed bacteria from scanning electron microscopy. Transcriptome sequencing results reveal that Ti-NTs-P-A screws interfered with ribosome formation and disrupted the arginine biosynthesis, which is crucial for bacterial survival in acidic environments. In the non-infected implant model, the bone-implant contact ratio of the Ti-NTs-P-A screw is 2.3 times that of the clinically used titanium screw. In an IASO model, Ti-NTs-P-A screws effectively eradicated bacteria within the OLCN, achieving an 80% infection control rate and desirable osteointegration. Collectively, Ti-NTs-P-A screws with pH-responsive antibacterial properties exhibit great potential for eradicating bacteria and achieving osseointegration in IASO.
Collapse
Affiliation(s)
- Hang Zhou
- Department of OrthopaedicsThe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400010P. R. China
| | - Youliang Ren
- Department of OrthopaedicsThe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400010P. R. China
| | - Kaixiong Zou
- National Engineering Research Center for Tissue Restoration and ReconstructionSouth China University of TechnologyGuangzhou510006P. R. China
| | - Ying Jin
- Department of OrthopaedicsThe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400010P. R. China
| | - Hang Liu
- Department of OrthopaedicsThe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400010P. R. China
| | - Haitao Jiang
- Department of OrthopaedicsThe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400010P. R. China
| | - Lei Shi
- Department of OrthopaedicsThe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400010P. R. China
| | - Xiaomin Sheng
- Department of OrthopaedicsThe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400010P. R. China
| | - Jason Weeks
- Department of OrthopaedicsNew York Medical CollegeNew YorkNY10595USA
| | - Hannah Wang
- Department of Orthopaedics, Center for Musculoskeletal ResearchUniversity of Rochester Medical CenterRochesterNY14642USA
| | - Thomas Xue
- Department of Orthopaedics, Center for Musculoskeletal ResearchUniversity of Rochester Medical CenterRochesterNY14642USA
| | - Edward M. Schwarz
- Department of Orthopaedics, Center for Musculoskeletal ResearchUniversity of Rochester Medical CenterRochesterNY14642USA
| | - Chao Xie
- Department of Orthopaedics, Center for Musculoskeletal ResearchUniversity of Rochester Medical CenterRochesterNY14642USA
| | - Zhongliang Deng
- Department of OrthopaedicsThe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400010P. R. China
| | - Lin Wang
- National Engineering Research Center for Tissue Restoration and ReconstructionSouth China University of TechnologyGuangzhou510006P. R. China
| | - Lei Chu
- Department of OrthopaedicsThe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400010P. R. China
| |
Collapse
|
17
|
Jagadale S, Damle M, Joshi MG. Bone Tissue Engineering: From Biomaterials to Clinical Trials. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025; 1479:73-115. [PMID: 39881051 DOI: 10.1007/5584_2024_841] [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: 01/31/2025]
Abstract
Bone tissue engineering is a promising field that aims to rebuild the bone tissue using biomaterials, cells, and signaling molecules. Materials like natural and synthetic polymers, inorganic materials, and composite materials are used to create scaffolds that mimic the hierarchical microstructure of bone. Stem cells, particularly mesenchymal stem cells (MSCs), play a crucial role in bone tissue engineering by promoting tissue regeneration and modulating the immune response. Growth factors like bone morphogenetic proteins (BMPs), vascular endothelial growth factor (VEGF), and platelet-derived growth factor (PDGF) are utilized to accelerate bone regeneration. Clinical applications include treating nonunion and mal-union fractures, osteonecrosis, orthopedic surgery, dental applications, and spinal cord injuries. Recent advances in the field include nanotechnology, 3D printing, bioprinting techniques, gene editing technologies, and microfluidic devices for drug testing. However, challenges remain, such as standardization of protocols, large-scale biomaterial production, personalized medicine approaches, cost-effectiveness, and regulatory issues. Current clinical trials are investigating the safety and efficacy of various bone tissue engineering approaches, with the potential to modernize patient care by providing more adequate treatments for bone defects and injuries.
Collapse
Affiliation(s)
- Swapnali Jagadale
- Department of Stem Cells & Regenerative Medicine, Centre for Interdisciplinary Research, D Y Patil Education Society (Deemed to be University), Kolhapur, India
| | - Mrunal Damle
- Department of Stem Cells & Regenerative Medicine, Centre for Interdisciplinary Research, D Y Patil Education Society (Deemed to be University), Kolhapur, India
| | - Meghnad G Joshi
- Department of Stem Cells & Regenerative Medicine, Centre for Interdisciplinary Research, D Y Patil Education Society (Deemed to be University), Kolhapur, India.
- Stem Plus Biotech, Sangli, India.
| |
Collapse
|
18
|
Zhao G, Zhao P, Wang Y, Zhang H, Zhu Y, Zhong J, You W, Shen G, Luo C, Mei O, Wu X, Li J, Shu Y, Wang H, Wagstaff W, Luu HH, Bi Y, Shi LL, Reid RR, He TC, Jiang L, Tang W, Fan J, Tang Z. GAPDH suppresses adenovirus-induced oxidative stress and enables a superfast production of recombinant adenovirus. Genes Dis 2024; 11:101344. [PMID: 39188753 PMCID: PMC11345542 DOI: 10.1016/j.gendis.2024.101344] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 05/15/2024] [Indexed: 08/28/2024] Open
Abstract
Recombinant adenovirus (rAdV) is a commonly used vector system for gene transfer. Efficient initial packaging and subsequent production of rAdV remains time-consuming and labor-intensive, possibly attributable to rAdV infection-associated oxidative stress and reactive oxygen species (ROS) production. Here, we show that exogenous GAPDH expression mitigates adenovirus-induced ROS-associated apoptosis in HEK293 cells, and expedites adenovirus production. By stably overexpressing GAPDH in HEK293 (293G) and 293pTP (293GP) cells, respectively, we demonstrated that rAdV-induced ROS production and cell apoptosis were significantly suppressed in 293G and 293GP cells. Transfection of 293G cells with adenoviral plasmid pAd-G2Luc yielded much higher titers of Ad-G2Luc at day 7 than that in HEK293 cells. Similarly, Ad-G2Luc was amplified more efficiently in 293G than in HEK293 cells. We further showed that transfection of 293GP cells with pAd-G2Luc produced much higher titers of Ad-G2Luc at day 5 than that of 293pTP cells. 293GP cells amplified the Ad-G2Luc much more efficiently than 293pTP cells, indicating that exogenous GAPDH can further augment pTP-enhanced adenovirus production. These results demonstrate that exogenous GAPDH can effectively suppress adenovirus-induced ROS and thus accelerate adenovirus production. Therefore, the engineered 293GP cells represent a superfast rAdV production system for adenovirus-based gene transfer and gene therapy.
Collapse
Affiliation(s)
- Guozhi Zhao
- Departments of Urology, Endocrinology, Orthopedic Surgery, and Gastroenterological Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Piao Zhao
- Departments of Urology, Endocrinology, Orthopedic Surgery, and Gastroenterological Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Ministry of Education Key Laboratory of Diagnostic Medicine, and Department of Clinical Biochemistry, School of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Yonghui Wang
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Department of Geriatrics, Xinhua Hospital, Shanghai Jiao-Tong University School of Medicine, Shanghai 200000, China
| | - Hui Zhang
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- The Breast Cancer Center, Chongqing University Cancer Hospital, Chongqing 4000430, China
| | - Yi Zhu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Department of Orthopaedic Surgery, Beijing Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| | - Jiamin Zhong
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Ministry of Education Key Laboratory of Diagnostic Medicine, and Department of Clinical Biochemistry, School of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Wulin You
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Department of Orthopaedic Surgery, Wuxi Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi, Jiangsu 214071, China
| | - Guowei Shen
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Department of Orthopaedic Surgery, Benq Medical Center, The Affiliated Benq Hospital of Nanjing Medical University, Nanjing, Jiangsu 210019, China
| | - Changqi Luo
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Department of Orthopaedic Surgery, Yibin Second People's Hospital, Affiliated with West China School of Medicine, Yibin, Sichuan 644000, China
| | - Ou Mei
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Department of Orthopedics, Jiangxi Hospital of Traditional Chinese Medicine, Jiangxi University of Traditional Chinese Medicine, Nanchang, Jiangxi 330006, China
| | - Xingye Wu
- Departments of Urology, Endocrinology, Orthopedic Surgery, and Gastroenterological Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Jingjing Li
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Department of Oncology, The Affiliated Hospital of Weifang Medical University, Weifang, Shandong 261053, China
| | - Yi Shu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Stem Cell Biology and Therapy Laboratory of the Pediatric Research Institute, The National Clinical Research Center for Child Health and Disorders, and Ministry of Education Key Laboratory of Child Development and Disorders, the Children's Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Hongwei Wang
- Division of Research and Development, Decoding Therapeutics, Inc., Mt Prospect, IL 60056, USA
| | - William Wagstaff
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Hue H. Luu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Yang Bi
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Stem Cell Biology and Therapy Laboratory of the Pediatric Research Institute, The National Clinical Research Center for Child Health and Disorders, and Ministry of Education Key Laboratory of Child Development and Disorders, the Children's Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Lewis L. Shi
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Russell R. Reid
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Laboratory of Craniofacial Biology and Development, Department of Surgery Section of Plastic Surgery, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Tong-Chuan He
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Laboratory of Craniofacial Biology and Development, Department of Surgery Section of Plastic Surgery, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Li Jiang
- Departments of Urology, Endocrinology, Orthopedic Surgery, and Gastroenterological Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Wei Tang
- Departments of Urology, Endocrinology, Orthopedic Surgery, and Gastroenterological Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Jiaming Fan
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Ministry of Education Key Laboratory of Diagnostic Medicine, and Department of Clinical Biochemistry, School of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Ziwei Tang
- Departments of Urology, Endocrinology, Orthopedic Surgery, and Gastroenterological Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| |
Collapse
|
19
|
Zhou J, Zhao Y, Ling Y, Zhao P, Gao H, Yang Y, Chen J. Microsphere-Composite Hydrogel for Recruiting Stem Cells and Promoting Osteogenic Differentiation. ACS APPLIED BIO MATERIALS 2024; 7:6520-6528. [PMID: 39263987 DOI: 10.1021/acsabm.4c00679] [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: 09/13/2024]
Abstract
By recruiting stem cells into scaffolds and differentiating them into osteoblasts, stem cells can be mobilized to directly repair bone defects, which avoids a series of disadvantages of exogenous stem cell implantation. In this study, a microsphere-composite hydrogel for the recruitment and osteogenic differentiation of stem cells was constructed. Methacrylic anhydride modified gelatin (GelMA) and heparin (HepMA), as well as nanohydroxyapatite (nHAP), were used to prepare microspheres followed by adsorbing platelet-derived growth factor BB (PDGF-BB) whose loading efficiency was 53.7 ± 2.2%. Then the microspheres were compounded to the GelMA hydrogel encapsulated with simvastatin (SIM) to obtain microsphere-composite hydrogel GHnH-P@GS. GHnH-P@GS hydrogel could slowly release SIM and PDGF-BB, and the extents of release within 7 days were 44.1 ± 2.0% and 32.8 ± 1.1%. The synergistic effect of small molecule drugs and growth factors not only induced the recruitment of rabbit bone marrow-derived mesenchymal stem cells, but also promoted the osteogenic differentiation of stem cells, which was confirmed by experiments of cell migration, alkaline phosphatase, and alizarin red staining. Collectively, the microsphere-composite hydrogel GHnH-P@GS has a certain reference significance for the design of scaffolds for alveolar bone repair and regeneration.
Collapse
Affiliation(s)
- Juan Zhou
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi 214122, China
| | - Yanling Zhao
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi 214122, China
| | - Yan Ling
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi 214122, China
| | - Peng Zhao
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi 214122, China
- Burn & Trauma Treatment Center, Affiliated Hospital of Jiangnan University, Wuxi 214122, China
| | - Hui Gao
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi 214122, China
| | - Yang Yang
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi 214122, China
- Burn & Trauma Treatment Center, Affiliated Hospital of Jiangnan University, Wuxi 214122, China
| | - Jinghua Chen
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi 214122, China
| |
Collapse
|
20
|
Cao B, Da X, Wu W, Xie J, Li X, Wang X, Xu H, Gao J, Yang H, Su J. Multifunctional human serum albumin-crosslinked and self-assembling nanoparticles for therapy of periodontitis by anti-oxidation, anti-inflammation and osteogenesis. Mater Today Bio 2024; 28:101163. [PMID: 39183771 PMCID: PMC11341939 DOI: 10.1016/j.mtbio.2024.101163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 07/02/2024] [Accepted: 07/18/2024] [Indexed: 08/27/2024] Open
Abstract
Periodontitis is a chronic inflammatory disease that can result in the irreversible loss of tooth-supporting tissues and elevate the likelihood and intensity of systemic diseases. The presence of reactive oxygen species (ROS) and associated related oxidative stress is intricately linked to the progression and severity of periodontal inflammation. Targeted removal of local ROS may serve to attenuate inflammation, improve the unfavorable periodontal microenvironment and potentially reverse ensuing pathological cascades. These ROS scavenging nanoparticles, which possess additional characteristics such as anti-inflammation and osteogenic differentiation, are highly sought after for the treatment of periodontitis. In this study, negative charged human serum albumin-crosslinked manganese-doped self-assembling Prussian blue nanoparticles (HSA-MDSPB NPs) were fabricated. These nanoparticles demonstrate the ability to scavenge multiple ROS including superoxide anion, free hydroxyl radicals, singlet oxygen and hydrogen peroxide. Additionally, HSA-MDSPB NPs exhibit the capacity to alleviate inflammation in gingiva and alveolar bone both in vitro and in vivo. Furthermore, HSA-MDSPB NPs have been shown to play a role in promoting the polarization of macrophages from the M1 to M2 phenotype, resulting in reduced production of pro-inflammatory cytokines. More attractively, HSA-MDSPB NPs have been demonstrated to enhance cellular osteogenic differentiation. These properties of HSA-MDSPB NPs contribute to decreased inflammation, extracellular matrix degradation and bone loss in periodontal tissue. In conclusion, the multifunctional nature of HSA-MDSPB NPs provides a promising therapeutic approach for the treatment of periodontitis.
Collapse
Affiliation(s)
- Bangping Cao
- Shanghai Engineering Research Center of Tooth Restoration and Regeneration & Tongji Research Institute of Stomatology & Department of Prosthodontics, Stomatological Hospital and Dental School, Tongji University, Shanghai, China
| | - Xuanbo Da
- Department of General Surgery, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710000, China
| | - Wenjing Wu
- Shanghai Engineering Research Center of Tooth Restoration and Regeneration & Tongji Research Institute of Stomatology & Department of Prosthodontics, Stomatological Hospital and Dental School, Tongji University, Shanghai, China
| | - Jian Xie
- Shanghai Engineering Research Center of Tooth Restoration and Regeneration & Tongji Research Institute of Stomatology & Department of Prosthodontics, Stomatological Hospital and Dental School, Tongji University, Shanghai, China
| | - Xuejing Li
- Shanghai Engineering Research Center of Tooth Restoration and Regeneration & Tongji Research Institute of Stomatology & Department of Prosthodontics, Stomatological Hospital and Dental School, Tongji University, Shanghai, China
| | - Xin Wang
- Shanghai Engineering Research Center of Tooth Restoration and Regeneration & Tongji Research Institute of Stomatology & Department of Prosthodontics, Stomatological Hospital and Dental School, Tongji University, Shanghai, China
| | - Hui Xu
- Shanghai Engineering Research Center of Tooth Restoration and Regeneration & Tongji Research Institute of Stomatology & Department of Prosthodontics, Stomatological Hospital and Dental School, Tongji University, Shanghai, China
| | - Jianfang Gao
- Shanghai Engineering Research Center of Tooth Restoration and Regeneration & Tongji Research Institute of Stomatology & Department of Prosthodontics, Stomatological Hospital and Dental School, Tongji University, Shanghai, China
| | - Hui Yang
- Shanghai Engineering Research Center of Tooth Restoration and Regeneration & Tongji Research Institute of Stomatology & Department of Prosthodontics, Stomatological Hospital and Dental School, Tongji University, Shanghai, China
| | - Jiansheng Su
- Shanghai Engineering Research Center of Tooth Restoration and Regeneration & Tongji Research Institute of Stomatology & Department of Prosthodontics, Stomatological Hospital and Dental School, Tongji University, Shanghai, China
| |
Collapse
|
21
|
Fan T, Zhu J, Liu W, Qu R, Khan AU, Shi Y, Liu J, Zhou Z, Xu C, Dai J, Ouyang J. SUN1 inhibits osteogenesis and promotes adipogenesis of human adipose-derived stem cells by regulating α-tubulin and CD36 expression. J Cell Mol Med 2024; 28:e70143. [PMID: 39383106 PMCID: PMC11463318 DOI: 10.1111/jcmm.70143] [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: 03/04/2024] [Revised: 08/29/2024] [Accepted: 10/01/2024] [Indexed: 10/11/2024] Open
Abstract
Sad and UNC84 domain 1 (SUN1) is a kind of nuclear envelope protein with established involvement in cellular processes, including nuclear motility and meiosis. SUN1 plays an intriguing role in human adipose-derived stem cells (hASCs) differentiation; however, this role remains largely undefined. This study was undertaken to investigate the role of SUN1 in hASCs differentiation, as well as its underlying mechanisms. Employing siRNAs, we selectively downregulated SUN1 and CD36 expression. Microtubules were depolymerized using nocodazole, and PPARγ was activated using rosiglitazone. Western blotting was performed to quantify SUN1, PPARγ, α-tubulin, CD36, OPN, and adiponectin protein expression levels. Alkaline phosphatase and Oil red O staining were used to assess osteogenesis and adipogenesis, respectively. Downregulated SUN1 expression increased osteogenesis and decreased adipogenesis in hASCs, concomitant with upregulated α-tubulin expression and downregulated CD36 expression, alongside reduced nuclear localization of PPARγ. Microtubule depolymerization increased CD36 expression. Rescue experiments indicated that microtubule depolymerization counteracted the downregulated SUN1-induced phenotypic changes. This study demonstrates that SUN1 influences the differentiation of hASCs towards osteogenic and adipogenic lineages, indicating its essential role in cell fate.
Collapse
Affiliation(s)
- Tingyu Fan
- Guangdong Provincial Key Laboratory of Digital Medicine and Biomechanics & Guangdong Engineering Research Center for Translation of Medical 3D Printing Application & National Virtual & Reality Experimental Education Center for Medical Morphology (Southern Medical University) & National Key Discipline of Human Anatomy, School of Basic Medical SciencesSouthern Medical UniversityGuangzhouChina
| | - Jinhui Zhu
- Guangdong Provincial Key Laboratory of Digital Medicine and Biomechanics & Guangdong Engineering Research Center for Translation of Medical 3D Printing Application & National Virtual & Reality Experimental Education Center for Medical Morphology (Southern Medical University) & National Key Discipline of Human Anatomy, School of Basic Medical SciencesSouthern Medical UniversityGuangzhouChina
| | - Wenqing Liu
- Guangdong Provincial Key Laboratory of Digital Medicine and Biomechanics & Guangdong Engineering Research Center for Translation of Medical 3D Printing Application & National Virtual & Reality Experimental Education Center for Medical Morphology (Southern Medical University) & National Key Discipline of Human Anatomy, School of Basic Medical SciencesSouthern Medical UniversityGuangzhouChina
| | - Rongmei Qu
- Guangdong Provincial Key Laboratory of Digital Medicine and Biomechanics & Guangdong Engineering Research Center for Translation of Medical 3D Printing Application & National Virtual & Reality Experimental Education Center for Medical Morphology (Southern Medical University) & National Key Discipline of Human Anatomy, School of Basic Medical SciencesSouthern Medical UniversityGuangzhouChina
| | - Asmat Ullah Khan
- Guangdong Provincial Key Laboratory of Digital Medicine and Biomechanics & Guangdong Engineering Research Center for Translation of Medical 3D Printing Application & National Virtual & Reality Experimental Education Center for Medical Morphology (Southern Medical University) & National Key Discipline of Human Anatomy, School of Basic Medical SciencesSouthern Medical UniversityGuangzhouChina
| | - Yulian Shi
- Guangdong Provincial Key Laboratory of Digital Medicine and Biomechanics & Guangdong Engineering Research Center for Translation of Medical 3D Printing Application & National Virtual & Reality Experimental Education Center for Medical Morphology (Southern Medical University) & National Key Discipline of Human Anatomy, School of Basic Medical SciencesSouthern Medical UniversityGuangzhouChina
| | - Jiaxuan Liu
- Guangdong Provincial Key Laboratory of Digital Medicine and Biomechanics & Guangdong Engineering Research Center for Translation of Medical 3D Printing Application & National Virtual & Reality Experimental Education Center for Medical Morphology (Southern Medical University) & National Key Discipline of Human Anatomy, School of Basic Medical SciencesSouthern Medical UniversityGuangzhouChina
| | - Zhitao Zhou
- Central LaboratorySouthern Medical UniversityGuangzhouChina
| | - Chujiang Xu
- Department of Orthopedics, TCM‐Integrated HospitalSouthern Medical UniversityGuangzhouChina
| | - Jingxing Dai
- Guangdong Provincial Key Laboratory of Digital Medicine and Biomechanics & Guangdong Engineering Research Center for Translation of Medical 3D Printing Application & National Virtual & Reality Experimental Education Center for Medical Morphology (Southern Medical University) & National Key Discipline of Human Anatomy, School of Basic Medical SciencesSouthern Medical UniversityGuangzhouChina
| | - Jun Ouyang
- Guangdong Provincial Key Laboratory of Digital Medicine and Biomechanics & Guangdong Engineering Research Center for Translation of Medical 3D Printing Application & National Virtual & Reality Experimental Education Center for Medical Morphology (Southern Medical University) & National Key Discipline of Human Anatomy, School of Basic Medical SciencesSouthern Medical UniversityGuangzhouChina
| |
Collapse
|
22
|
Wu E, Huang L, Shen Y, Wei Z, Li Y, Wang J, Chen Z. Application of gelatin-based composites in bone tissue engineering. Heliyon 2024; 10:e36258. [PMID: 39224337 PMCID: PMC11367464 DOI: 10.1016/j.heliyon.2024.e36258] [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: 04/01/2024] [Revised: 07/25/2024] [Accepted: 08/13/2024] [Indexed: 09/04/2024] Open
Abstract
Natural bone tissue has the certain function of self-regeneration and repair, but it is difficult to repair large bone damage. Recently, although autologous bone grafting is the "gold standard" for improving bone repair, it has high cost, few donor sources. Besides, allogeneic bone grafting causes greater immune reactions, which hardly meet clinical needs. The bone tissue engineering (BTE) has been developed to promote bone repair. Gelatin, due to its biocompatibility, receives a great deal of attention in the BTE research field. However, the disadvantages of natural gelatin are poor mechanical properties and single structural property. With the development of BTE, gelatin is often used in combination with a range of natural, synthetic polymers, and inorganic materials to achieve synergistic effects for the complex physiological process of bone repair. The review delves into the fundamental structure and unique properties of gelatin, as well as the excellent properties necessary for bone scaffold materials. Then this review explores the application of modified gelatin three-dimensional (3D) scaffolds with various structures in bone repair, including 3D fiber scaffolds, hydrogels, and nanoparticles. In addition, the review focuses on the excellent efficacy of composite bone tissue scaffolds consisting of modified gelatin, various natural or synthetic polymeric materials, as well as bioactive ceramics and inorganic metallic/non-metallic materials in the repair of bone defects. The combination of these gelatin-based composite scaffolds provides new ideas for the design of scaffold materials for bone repair with good biosafety.
Collapse
Affiliation(s)
- Enguang Wu
- Jiangxi Province Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science and Technology Normal University, Nanchang, 330013, China
| | - Lianghui Huang
- Jiangxi Province Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science and Technology Normal University, Nanchang, 330013, China
| | - Yao Shen
- Jiangxi Province Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science and Technology Normal University, Nanchang, 330013, China
| | - Zongyi Wei
- Jiangxi Province Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science and Technology Normal University, Nanchang, 330013, China
| | - Yangbiao Li
- Jiangxi Province Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science and Technology Normal University, Nanchang, 330013, China
| | - Jin Wang
- Jiangxi Province Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science and Technology Normal University, Nanchang, 330013, China
| | - Zhenhua Chen
- Jiangxi Province Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science and Technology Normal University, Nanchang, 330013, China
| |
Collapse
|
23
|
Zhu Y, Mei O, Zhang H, You W, Zhong J, Collins CP, Shen G, Luo C, Wu X, Li J, Shu Y, Wen Y, Luu HH, Shi LL, Fan J, He TC, Ameer GA, Sun C, Wen L, Reid RR. Establishment and characterization of a rat model of scalp-cranial composite defect for multilayered tissue engineering. RESEARCH SQUARE 2024:rs.3.rs-4643966. [PMID: 39108474 PMCID: PMC11302684 DOI: 10.21203/rs.3.rs-4643966/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 08/12/2024]
Abstract
Composite cranial defects have individual functional and aesthetic ramifications, as well as societal burden, while posing significant challenges for reconstructive surgeons. Single-stage composite reconstruction of these deformities entail complex surgeries that bear many short- and long-term risks and complications. Current research on composite scalp-cranial defects is sparse and one-dimensional, often focusing solely on bone or skin. Thus, there is an unmet need for a simple, clinically relevant composite defect model in rodents, where there is a challenge in averting healing of the skin component via secondary intention. By utilizing a customizable (3D-printed) wound obturator, the scalp wound can be rendered non-healing for a long period (more than 6 weeks), with the cranial defect patent. The wound obturator shows minimal biotoxicity and will not cause severe endocranium-granulation adhesion. This composite defect model effectively slowed the scalp healing process and preserved the cranial defect, embodying the characteristics of a "chronic composite defect". In parallel, an autologous reconstruction model was established as the positive control. This positive control exhibited reproducible healing of the skin within 3 weeks with variable degrees of osseointegration, consistent with clinical practice. Both models provide a stable platform for subsequent research not only for composite tissue engineering and scaffold design but also for mechanistic studies of composite tissue healing.
Collapse
Affiliation(s)
- Yi Zhu
- The University of Chicago Medical Center
| | - Ou Mei
- The University of Chicago Medical Center
| | - Hui Zhang
- The University of Chicago Medical Center
| | - Wulin You
- The University of Chicago Medical Center
| | | | | | | | | | - Xingye Wu
- The University of Chicago Medical Center
| | | | - Yi Shu
- The University of Chicago Medical Center
| | - Ya Wen
- Capital Medical University
| | - Hue H Luu
- The University of Chicago Medical Center
| | | | | | | | | | | | - Liangyuan Wen
- Chinese Academy of Medical Sciences & Peking Union Medical College
| | | |
Collapse
|
24
|
柯 志, 黄 子, 何 若, 张 倩, 陈 思, 崔 忠, 丁 晶. [ Hmga2 knockdown enhances osteogenic differentiation of adipose-derived mesenchymal stem cells and accelerates bone defect healing in mice]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2024; 44:1227-1235. [PMID: 39051068 PMCID: PMC11270651 DOI: 10.12122/j.issn.1673-4254.2024.07.02] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Indexed: 07/27/2024]
Abstract
OBJECTIVE To investigate the role of high-mobility group AT-hook 2 (HMGA2) in osteogenic differentiation of adipose-derived mesenchymal stem cells (ADSCs) and the effect of Hmga2 knockdown for promoting bone defect repair. METHODS Bioinformatics studies using the GEO database and Rstudio software identified HMGA2 as a key factor in adipogenic-osteogenic differentiation balance of ADSCs. The protein-protein interaction network of HMGA2 in osteogenic differentiation was mapped using String and visualized with Cytoscape to predict the downstream targets of HMGA2. Primary mouse ADSCs (mADSCs) were transfected with Hmga2 siRNA, and the changes in osteogenic differentiation of the cells were evaluated using alkaline phosphatase staining and Alizarin red S staining. The expressions of osteogenic markers Runt-related transcription factor 2 (RUNX2), osteopontin (OPN), and osteocalcein (OCN) in the transfected cells were detected using RT-qPCR and Western blotting. In a mouse model of critical-sized calvarial defects, mADSCs with Hmga2-knockdown were transplanted into the defect, and bone repair was evaluated 6 weeks later using micro-CT scanning and histological staining. RESULTS GEO database analysis showed that HMGA2 expression was upregulated during adipogenic differentiation of ADSCs. Protein-protein interaction network analysis suggested that the potential HMGA2 targets in osteogenic differentiation of ADSCs included SMAD7, CDH1, CDH2, SNAI1, SMAD9, IGF2BP3, and ALDH1A1. In mADSCs, Hmga2 knockdown significantly upregulated the expressions of RUNX2, OPN, and OCN and increased cellular alkaline phosphatase activity and calcium deposition. In a critical-sized calvarial defect model, transplantation of mADSCs with Hmga2 knockdown significantly promoted new bone formation. CONCLUSION HMGA2 is a crucial regulator of osteogenic differentiation in ADSCs, and Hmga2 knockdown significantly promotes osteogenic differentiation of ADSCs and accelerates ADSCs-mediated bone defect repair in mice.
Collapse
|
25
|
Alqasoumi A, Alsharidah M, Mahmood A, Elsafadi M, Al Rugaie O, Mohany KM, Al-Regaiey KA, Alyahya KI, Alanteet AA, Algarzae NK, AlGhibiwi HK, AlHomaidi A, Abumaree M. Mesenchymal Stem Cell-Conditioned Media Modulate HUVEC Response to H 2O 2: Impact on Gene Expression and Potential for Atherosclerosis Intervention. BIOMED RESEARCH INTERNATIONAL 2024; 2024:7726493. [PMID: 39050921 PMCID: PMC11268959 DOI: 10.1155/2024/7726493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 05/31/2024] [Accepted: 06/13/2024] [Indexed: 07/27/2024]
Abstract
Background: We studied the potential of human bone marrow-derived mesenchymal stem cell conditioned media (hBMSC CM) in protecting endothelial cell properties (viability, proliferation, and migrations) from the deleterious effects produced by the inflammatory environment of H2O2. Additionally, we investigated their impact on the endothelial cells' gene expression of some inflammatory-related genes, namely, TGF-β1, FOS, ATF3, RAF-1, and SMAD3. Methods: Human umbilical vein endothelial cells (HUVECs) were cultured individually under three conditions: alone, with varying concentrations of H2O2, or with varying concentrations of H2O2 and hBMSC CM. HUVEC adhesion, proliferation, and migration were evaluated using the xCELLigence system. The HUVECs' gene expressions were evaluated by real-time polymerase chain reaction (RT-PCR). Results: Generally, we observed enhanced HUVEC viability, proliferation, and migration when cultured in media supplemented with H2O2 and hBMSC CM. Furthermore, the CM modulated the expressions of the studied inflammatory-related genes in HUVECs, promoting a more robust cellular response. Conclusion: This study has illuminated the protective role of hBMSC CM in mitigating the damaging effects of H2O2 on endothelial cell function. Our data demonstrate that hBMSC CM enhances the viability, proliferation, and migration of HUVECs even under oxidative stress conditions. Additionally, the conditioned medium was found to modulate the gene expression of pivotal markers related to inflammation, suggesting a favorable influence on cellular response mechanisms.
Collapse
Affiliation(s)
- Abdulmajeed Alqasoumi
- Department of Pharmacy PracticeCollege of PharmacyQassim University, Qassim, Saudi Arabia
| | - Mansour Alsharidah
- Department of PhysiologyCollege of MedicineQassim University, Qassim, Saudi Arabia
| | - Amer Mahmood
- Stem Cell Unit Department of AnatomyCollege of MedicineKing Saud University, Riyadh, Saudi Arabia
| | - Mona Elsafadi
- Stem Cell Unit Department of AnatomyCollege of MedicineKing Saud University, Riyadh, Saudi Arabia
| | - Osamah Al Rugaie
- Department of Biology and ImmunologyCollege of MedicineQassim University, Qassim, Saudi Arabia
| | - Khalid M. Mohany
- Department of Medical BiochemistryFaculty of MedicineAssiut University, El Gamma Street, Assiut City 71515, Egypt
| | - Khalid A. Al-Regaiey
- Department of PhysiologyCollege of MedicineKing Saud University, Riyadh, Saudi Arabia
| | - Khaleel I. Alyahya
- Department of AnatomyCollege of MedicineKing Saud University, Riyadh, Saudi Arabia
| | - Alaa A. Alanteet
- Department of Pharmacology and ToxicologyCollege of PharmacyKing Saud University, Riyadh, Saudi Arabia
| | - Norah K. Algarzae
- Department of PhysiologyCollege of MedicineKing Saud University, Riyadh, Saudi Arabia
| | - Hanan K. AlGhibiwi
- Department of Pharmacology and ToxicologyCollege of PharmacyKing Saud University, Riyadh, Saudi Arabia
| | - Adel AlHomaidi
- Department of PathologyCollege of MedicineQassim University, Qassim, Saudi Arabia
| | - Mohammad Abumaree
- Stem Cells and Regenerative MedicineCell Therapy and Cancer Research (CTCR)King Abdullah International Medical Research Center (KAIMRC)King Saud Bin Abdulaziz University for Health Sciences (KSAU)King Abdulaziz Medical CityMinistry of National Guard Health Affairs (MNGHA), Riyadh, Saudi Arabia
| |
Collapse
|
26
|
Waqar MA, Zaman M, Khan R, Shafeeq Ur Rahman M, Majeed I. Navigating the tumor microenvironment: mesenchymal stem cell-mediated delivery of anticancer agents. J Drug Target 2024; 32:624-634. [PMID: 38652480 DOI: 10.1080/1061186x.2024.2347356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 04/21/2024] [Indexed: 04/25/2024]
Abstract
Scientific knowledge of cancer has advanced greatly throughout the years, with most recent studies findings includes many hallmarks that capture disease's multifaceted character. One of the novel approach utilised for the delivery of anti-cancer agents includes mesenchymal stem cell mediated drug delivery. Mesenchymal stem cells (MSCs) are non-haematopoietic progenitor cells that may be extracted from bone marrow, tooth pulp, adipose tissue and placenta/umbilical cord blood dealing with adult stem cells. MSCs are mostly involved in regeneration of tissue, they have also been shown to preferentially migrate to location of several types of tumour in-vivo. Usage of MSCs ought to improve both effectiveness and safety of anti-cancer drugs by enhancing delivery efficiency of anti-cancer therapies to tumour site. Numerous researches has demonstrated that various drugs, when delivered via mesenchymal stem cell mediated delivery can elicit anti-tumour effect of cells in cancers of breast cells and thyroid cells. MSCs have minimal immunogenicity because to lack of co-stimulatory molecule expression, which means there is no requirement for immunosuppression after allogenic transplantation. This current review elaborates recent advancements of mesenchyma stem cell mediated drug delivery of anti-cancer agents along with its mechanism and previously reported studies of drugs manufactured via this drug delivery system.
Collapse
Affiliation(s)
- Muhammad Ahsan Waqar
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Lahore University of Biological & Applied Sciences, Lahore, Pakistan
| | - Muhammad Zaman
- Faculty of Pharmaceutical Sciences, University of Central Punjab, Lahore, Pakistan
| | - Rabeel Khan
- Department of Pharmacy Practice, Faculty of Pharmaceutical Sciences, Lahore University of Biological & Applied Sciences, Lahore, Pakistan
| | | | - Imtiaz Majeed
- Faculty of Pharmaceutical Sciences, University of Central Punjab, Lahore, Pakistan
| |
Collapse
|
27
|
Zhou H, He Z, Cao Y, Chu L, Liang B, Yu K, Deng Z. An injectable magnesium-loaded hydrogel releases hydrogen to promote osteoporotic bone repair via ROS scavenging and immunomodulation. Theranostics 2024; 14:3739-3759. [PMID: 38948054 PMCID: PMC11209720 DOI: 10.7150/thno.97412] [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: 04/17/2024] [Accepted: 06/03/2024] [Indexed: 07/02/2024] Open
Abstract
Background: The repair of osteoporotic bone defects remains challenging due to excessive reactive oxygen species (ROS), persistent inflammation, and an imbalance between osteogenesis and osteoclastogenesis. Methods: Here, an injectable H2-releasing hydrogel (magnesium@polyethylene glycol-poly(lactic-co-glycolic acid), Mg@PEG-PLGA) was developed to remodel the challenging bone environment and accelerate the repair of osteoporotic bone defects. Results: This Mg@PEG-PLGA gel shows excellent injectability, shape adaptability, and phase-transition ability, can fill irregular bone defect areas via minimally invasive injection, and can transform into a porous scaffold in situ to provide mechanical support. With the appropriate release of H2 and magnesium ions, the 2Mg@PEG-PLGA gel (loaded with 2 mg of Mg) displayed significant immunomodulatory effects through reducing intracellular ROS, guiding macrophage polarization toward the M2 phenotype, and inhibiting the IκB/NF-κB signaling pathway. Moreover, in vitro experiments showed that the 2Mg@PEG-PLGA gel inhibited osteoclastogenesis while promoting osteogenesis. Most notably, in animal experiments, the 2Mg@PEG-PLGA gel significantly promoted the repair of osteoporotic bone defects in vivo by scavenging ROS and inhibiting inflammation and osteoclastogenesis. Conclusions: Overall, our study provides critical insight into the design and development of H2-releasing magnesium-based hydrogels as potential implants for repairing osteoporotic bone defects.
Collapse
Affiliation(s)
- Hang Zhou
- Department of Orthopaedics, the Second Affiliated Hospital of Chongqing Medical University, 76 Linjiang Road, Yuzhong Distinct, Chongqing, 400010, P. R. China
- Department of Ultrasound & Chongqing Key Laboratory of Ultrasound Molecular Imaging, the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P. R. China
| | - Zhongyuan He
- Department of Orthopaedics, the Second Affiliated Hospital of Chongqing Medical University, 76 Linjiang Road, Yuzhong Distinct, Chongqing, 400010, P. R. China
| | - Youde Cao
- Department of Pathology from College of Basic Medicine, and Molecular Medicine Diagnostic & Testing Center, and Department of Clinical Pathology Laboratory of Pathology Diagnostic Center, Chongqing Medical University, 1 Yixueyuan Road, Yuzhong Distinct, Chongqing 400016, P. R. China
| | - Lei Chu
- Department of Orthopaedics, the Second Affiliated Hospital of Chongqing Medical University, 76 Linjiang Road, Yuzhong Distinct, Chongqing, 400010, P. R. China
| | - Bing Liang
- Department of Pathology from College of Basic Medicine, and Molecular Medicine Diagnostic & Testing Center, and Department of Clinical Pathology Laboratory of Pathology Diagnostic Center, Chongqing Medical University, 1 Yixueyuan Road, Yuzhong Distinct, Chongqing 400016, P. R. China
| | - Kexiao Yu
- Department of Orthopedics, Chongqing Traditional Chinese Medicine Hospital, The First Affiliated Hospital of Chongqing College of Traditional Chinese Medicine, No. 6 Panxi Seventh Branch Road, Jiangbei District, Chongqing 400021, P. R. China
| | - Zhongliang Deng
- Department of Orthopaedics, the Second Affiliated Hospital of Chongqing Medical University, 76 Linjiang Road, Yuzhong Distinct, Chongqing, 400010, P. R. China
| |
Collapse
|
28
|
Astaneh ME, Noori F, Fereydouni N. Curcumin-loaded scaffolds in bone regeneration. Heliyon 2024; 10:e32566. [PMID: 38961905 PMCID: PMC11219509 DOI: 10.1016/j.heliyon.2024.e32566] [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: 12/27/2023] [Revised: 06/05/2024] [Accepted: 06/05/2024] [Indexed: 07/05/2024] Open
Abstract
In recent years, there has been a notable surge in the development of engineered bone scaffolds intended for the repair of bone defects. While autografts and allografts have traditionally served as the primary methods in bone tissue engineering, their inherent limitations have spurred the exploration of novel avenues in biomedical implant development. The emergence of bone scaffolds not only facilitates bone reconstruction but also offers a platform for the targeted delivery of therapeutic agents. There exists a pervasive interest in leveraging various drugs, proteins, growth factors, and biomolecules with osteogenic properties to augment bone formation, as the enduring side effects associated with current clinical modalities necessitate the pursuit of safer alternatives. Curcumin, the principal bioactive compound found in turmeric, has demonstrated notable efficacy in regulating the proliferation and differentiation of bone cells while promoting bone formation. Nevertheless, its utility is hindered by restricted water solubility and poor bioavailability. Strategies aimed at enhancing the solubility, stability, and bioavailability of curcumin, including formulation techniques such as liposomes and nanoparticles or its complexation with metals, have been explored. This investigation is dedicated to exploring the impact of curcumin on the proliferation, differentiation, and migration of osteocytes, osteoblasts, and osteoclasts.
Collapse
Affiliation(s)
- Mohammad Ebrahim Astaneh
- Department of Anatomical Sciences, School of Medicine, Fasa University of Medical Sciences, Fasa, Iran
- Department of Tissue Engineering, School of Medicine, Fasa University of Medical Sciences, Fasa, Iran
- Student Research Committee, Fasa University of Medical Sciences, Fasa, Iran
| | - Fariba Noori
- Department of Tissue Engineering, School of Medicine, Fasa University of Medical Sciences, Fasa, Iran
- Student Research Committee, Fasa University of Medical Sciences, Fasa, Iran
| | - Narges Fereydouni
- Department of Tissue Engineering, School of Medicine, Fasa University of Medical Sciences, Fasa, Iran
- Student Research Committee, Fasa University of Medical Sciences, Fasa, Iran
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran
| |
Collapse
|
29
|
Emami Meybodi SM, Moradi Moraddahande F, Dehghani Firoozabadi A. Immunogenic cell death mediated TLR3/4-activated MSCs in U87 GBM cell line. Heliyon 2024; 10:e29858. [PMID: 38698968 PMCID: PMC11064142 DOI: 10.1016/j.heliyon.2024.e29858] [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: 02/17/2024] [Revised: 04/15/2024] [Accepted: 04/16/2024] [Indexed: 05/05/2024] Open
Abstract
Background and aims Glioblastoma (GBM) is an aggressive primary brain cancer with no promising curative therapies. It has been indicated that MSCs can interact with the tumour microenvironment (TME) through the secretion of soluble mediators regulating intercellular signalling within the TME. TLRs are a multigene family of pattern recognition receptors with evolutionarily conserved regions and are widely expressed in immune and other body cells. MSCs by TLRs can recognize conserved molecular components (DAPMPs and PAPMPs) and activate signalling pathways, which regulate immune and inflammatory responses. MSCs may exert immunomodulatory functions through interaction with their expressed toll-like receptors (TLRs) and exert a protective effect against tumour antigens. As an emerging approach, we aimed to monitor the U87 cell line growth, migration and death markers following specific TLR3/4-primed-MSCs-CMs treatment. Methods and results We investigated the phenotypic and functional outcomes of primed-CMs and glioma cell line co-culture following short-term, low-dose TLR3/4 priming. The gene expression profile of target genes, including apoptotic markers and related genes, was analyzed by qRT-PCR. MicroRNA-Seq examined the miRNA expression patterns, and flow cytometry evaluated the cell viability and cycle stages. The results showed significant changes in apoptosis and likely necroptosis-related markers following TLR3/4-primed-MSCs-CMs exposure in the glioma cell line. Notably, we observed a considerable induction of selective pro-apoptotic markers and both the early and late stages of apoptosis in treated U87 cell lines. Additionally, the migration rate of glioma cells significantly decreased following MSCs-CM treatment. Conclusion Our findings confirmed that the exposure of TLR3/4-activated-MSCs-CMs with glioma tumour cells possibly changes the immunogenicity of the tumour microenvironment and induces immunogenic programmed cell death. Our results can support the idea that TLR3/4-primed-MSCs can lead to innate immune-mediated cell death and modify tumour cell biology in invasive and metastatic cancers.
Collapse
Affiliation(s)
- Seyed Mahdi Emami Meybodi
- Yazd Cardiovascular Research Center, Non-Communicable Diseases Research Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Fateme Moradi Moraddahande
- Department of Medical Laboratory Sciences, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ali Dehghani Firoozabadi
- Yazd Cardiovascular Research Center, Non-Communicable Diseases Research Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| |
Collapse
|
30
|
Lian M, Qiao Z, Qiao S, Zhang X, Lin J, Xu R, Zhu N, Tang T, Huang Z, Jiang W, Shi J, Hao Y, Lai H, Dai K. Nerve Growth Factor-Preconditioned Mesenchymal Stem Cell-Derived Exosome-Functionalized 3D-Printed Hierarchical Porous Scaffolds with Neuro-Promotive Properties for Enhancing Innervated Bone Regeneration. ACS NANO 2024; 18:7504-7520. [PMID: 38412232 DOI: 10.1021/acsnano.3c11890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/29/2024]
Abstract
The essential role of the neural network in enhancing bone regeneration has often been overlooked in biomaterial design, leading to delayed or compromised bone healing. Engineered mesenchymal stem cells (MSCs)-derived exosomes are becoming increasingly recognized as potent cell-free agents for manipulating cellular behavior and improving therapeutic effectiveness. Herein, MSCs are stimulated with nerve growth factor (NGF) to regulate exosomal cargoes to improve neuro-promotive potential and facilitate innervated bone regeneration. In vitro cell experiments showed that the NGF-stimulated MSCs-derived exosomes (N-Exos) obviously improved the cellular function and neurotrophic effects of the neural cells, and consequently, the osteogenic potential of the osteo-reparative cells. Bioinformatic analysis by miRNA sequencing and pathway enrichment revealed that the beneficial effects of N-Exos may partly be ascribed to the NGF-elicited multicomponent exosomal miRNAs and the subsequent regulation and activation of the MAPK and PI3K-Akt signaling pathways. On this basis, N-Exos were delivered on the micropores of the 3D-printed hierarchical porous scaffold to accomplish the sustained release profile and extended bioavailability. In a rat model with a distal femoral defect, the N-Exos-functionalized hierarchical porous scaffold significantly induced neurovascular structure formation and innervated bone regeneration. This study provided a feasible strategy to modulate the functional cargoes of MSCs-derived exosomes to acquire desirable neuro-promotive and osteogenic potential. Furthermore, the developed N-Exos-functionalized hierarchical porous scaffold may represent a promising neurovascular-promotive bone reparative scaffold for clinical translation.
Collapse
Affiliation(s)
- Meifei Lian
- Department of Oral and Maxillofacial Implantology, Shanghai PerioImplant Innovation Center, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai 200011, China
- Clinical and Translational Research Center for 3D Printing Technology, Medical 3D Printing Innovation Research Center, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200125, China
| | - Zhiguang Qiao
- Clinical and Translational Research Center for 3D Printing Technology, Medical 3D Printing Innovation Research Center, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200125, China
- Department of Bone and Joint Surgery, Department of Orthopedics, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200001, China
| | - Shichong Qiao
- Department of Oral and Maxillofacial Implantology, Shanghai PerioImplant Innovation Center, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai 200011, China
| | - Xing Zhang
- State Key Laboratory of Mechanical Systems and Vibration, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Jieming Lin
- Department of Bone and Joint Surgery, Department of Orthopedics, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200001, China
| | - Ruida Xu
- Department of Bone and Joint Surgery, Department of Orthopedics, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200001, China
| | - Naifeng Zhu
- Department of Bone and Joint Surgery, Department of Orthopedics, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200001, China
| | - Tianhong Tang
- Department of Prosthodontics, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Zhuoli Huang
- Department of Oral and Maxillofacial Implantology, Shanghai PerioImplant Innovation Center, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai 200011, China
| | - Wenbo Jiang
- Clinical and Translational Research Center for 3D Printing Technology, Medical 3D Printing Innovation Research Center, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200125, China
| | - Junyu Shi
- Department of Oral and Maxillofacial Implantology, Shanghai PerioImplant Innovation Center, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai 200011, China
| | - Yongqiang Hao
- Clinical and Translational Research Center for 3D Printing Technology, Medical 3D Printing Innovation Research Center, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200125, China
- Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Hongchang Lai
- Department of Oral and Maxillofacial Implantology, Shanghai PerioImplant Innovation Center, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai 200011, China
| | - Kerong Dai
- Clinical and Translational Research Center for 3D Printing Technology, Medical 3D Printing Innovation Research Center, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200125, China
- Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| |
Collapse
|