1
|
Mi Y, Wei D, Du B, Zhang R, Li J, Huang S, Zhang B, Ren J, Wu X. Effect of type 2 diabetes mellitus microenvironment on osteogenic capacity of bone marrow mesenchymal stem cells. Int Immunopharmacol 2025; 157:114724. [PMID: 40300360 DOI: 10.1016/j.intimp.2025.114724] [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: 02/27/2025] [Revised: 04/07/2025] [Accepted: 04/21/2025] [Indexed: 05/01/2025]
Abstract
Type 2 diabetes mellitus (T2DM) often leads to delayed bone regeneration such as slow healing of fractures and bone defects. The number, status and osteogenic differentiation capacity of bone marrow mesenchymal stem cells (BMSCs) are extremely important in bone healing and bone regeneration. The T2DM microenvironment can have irreversible negative effects on BMSCs. In this paper, we review the molecular expression and altered proliferation, migration, and osteogenic differentiation capacity of BMSCs in the microenvironment of T2DM, it provides a new perspective to restore the normal function of T2DM-BMSCs, so as to save the damaged bone regeneration capacity.
Collapse
Affiliation(s)
- Yanling Mi
- Shanxi Medical University, School and Hospital of Stomatology, Taiyuan 030001, China; Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan 030001, China
| | - Danni Wei
- Shanxi Medical University, School and Hospital of Stomatology, Taiyuan 030001, China; Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan 030001, China
| | - Bingli Du
- Shanxi Medical University, School and Hospital of Stomatology, Taiyuan 030001, China; Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan 030001, China
| | - Ran Zhang
- Shanxi Medical University, School and Hospital of Stomatology, Taiyuan 030001, China; Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan 030001, China
| | - Jiadi Li
- Shanxi Medical University, School and Hospital of Stomatology, Taiyuan 030001, China; Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan 030001, China
| | - Shuo Huang
- Shanxi Medical University, School and Hospital of Stomatology, Taiyuan 030001, China; Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan 030001, China
| | - Binbin Zhang
- Shanxi Medical University, School and Hospital of Stomatology, Taiyuan 030001, China; Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan 030001, China
| | - Juan Ren
- Shanxi Medical University, School and Hospital of Stomatology, Taiyuan 030001, China; Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan 030001, China.
| | - Xiuping Wu
- Shanxi Medical University, School and Hospital of Stomatology, Taiyuan 030001, China; Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan 030001, China.
| |
Collapse
|
2
|
de Morais PB, de Almeida GS, de Camargo Andrade AF, Orsi RDO, Zambuzzi WF, Fernandes CJDC. Modulation of HIF-1α and TNF-α in pre-osteoblasts treated with alcohol extract of propolis: Implications for cellular response and signaling pathways. Tissue Cell 2025; 94:102784. [PMID: 39987775 DOI: 10.1016/j.tice.2025.102784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 01/28/2025] [Accepted: 02/03/2025] [Indexed: 02/25/2025]
Abstract
Numerous studies have demonstrated the significant role of propolis in various biological processes, including inflammation and the repair of endothelial and bone tissues. The cascade of inflammation, proliferation, and tissue remodeling characterizes the injury site. Remarkable progress has been made in the field of alternative medicine with the utilization of propolis. In this study, we specifically investigated the effects of alcoholic extract of propolis on cell adhesion, survival, remodeling, and differentiation pathways in osteoblasts. Our findings revealed the activation of survival proteins such as AKT, ERK, and phosphorylated ERK during the adhesion phase, while interleukins, specifically IL-6 and TNF, exhibited increased expression during cell differentiation. Furthermore, treatment with propolis extract led to enhanced activity of metalloproteinases (MMP9 and MMP2) and HIF-1α, a crucial activator of angiogenesis and bone remodeling. It was observed that the alcoholic propolis extract stimulated cell proliferation, differentiation, and repair, with key molecules involved in these processes including pro-inflammatory factors IL-6 and TNFα, as well as HIF-1, MMP2, and BMP7 proteins. Collectively, these factors induced an initial state of proliferation followed by differentiation and repair. Therefore, the alcoholic extract of propolis holds promise as a potential therapeutic agent, particularly in cases involving tissue repair. Nevertheless, further comprehensive in vitro and in vivo studies targeting this area are necessary to advance our understanding.
Collapse
Affiliation(s)
- Paula Bertin de Morais
- Lab. of Bioassays and Cell Dynamics, Department of Chemical and Biological Sciences, Institute of Biosciences, Universidade Estadual Paulista - UNESP, Botucatu, São Paulo CEP 18618-970, Brazil
| | - Gerson Santos de Almeida
- Lab. of Bioassays and Cell Dynamics, Department of Chemical and Biological Sciences, Institute of Biosciences, Universidade Estadual Paulista - UNESP, Botucatu, São Paulo CEP 18618-970, Brazil
| | - Amanda Fantini de Camargo Andrade
- Lab. of Bioassays and Cell Dynamics, Department of Chemical and Biological Sciences, Institute of Biosciences, Universidade Estadual Paulista - UNESP, Botucatu, São Paulo CEP 18618-970, Brazil
| | - Ricardo de Oliveira Orsi
- Department of Animal Production and Exploration, Faculdade de Medicina Veterinária e Zootecnia, Universidade Estadual Paulista - UNESP, Botucatu, São Paulo CEP 18618-681, Brazil
| | - Willian Fernando Zambuzzi
- Lab. of Bioassays and Cell Dynamics, Department of Chemical and Biological Sciences, Institute of Biosciences, Universidade Estadual Paulista - UNESP, Botucatu, São Paulo CEP 18618-970, Brazil
| | - Célio Jr Da Costa Fernandes
- Lab. of Bioassays and Cell Dynamics, Department of Chemical and Biological Sciences, Institute of Biosciences, Universidade Estadual Paulista - UNESP, Botucatu, São Paulo CEP 18618-970, Brazil; Department of Biophysics and Phamacology, Institut of Biosciences, Universidade Estadual Paulista - UNESP, Brazil.
| |
Collapse
|
3
|
Yu J, Yang M, Jin Y, Yang K, Yang H. Insights into the potential role of BMSCs-exo delivered USP14 on SIRT1 deubiquitination in Staphylococcus aureus-induced model of osteomyelitis. J Orthop Surg Res 2025; 20:539. [PMID: 40442730 PMCID: PMC12120999 DOI: 10.1186/s13018-025-05898-7] [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: 12/25/2024] [Accepted: 05/06/2025] [Indexed: 06/02/2025] Open
Abstract
Osteomyelitis resulting from a traumatic fracture is a recurrent and difficult-to-treat bone infection. Ubiquitin-specific protease 14 (USP14), a deubiquitinating enzyme, and Sirtuin-1 (SIRT1), an NAD+-dependent deacetylase, both play critical roles in regulating cellular processes, including inflammation. It has been discovered that exosomes originated from bone marrow mesenchymal stem cells (BMSCs-exo) can promote the repair and regeneration of bone fractures. In this study, we aimed to investigate the role of BMSCs-exo in osteoblast differentiation in osteomyelitis and the related molecular mechanisms. MC3T3-E1 cells induced with S. aureus were used as an in vitro model of osteomyelitis. BMSCs-exo were isolated and characterized using ultracentrifugation, transmission electron microscopy (TEM), and Western blot. RT-qPCR, Western blot, CCK-8, ALP staining, ELISA, and CO-IP were utilized to evaluate USP14 and SIRT1 levels, the osteogenic differentiation ability of MC3T3-E1 cells, and the deubiquitination level of SIRT1. Low expression of USP14 and SIRT1 was observed in the bone tissue of osteomyelitis patients. BMSCs-exo could upregulate the expression of USP14 and promote the expression of SIRT1 protein in the cell model of osteomyelitis. In addition, BMSCs-exo reduced the levels of inflammatory factors TNFα and IL-6, enhanced cell viability, promoted the expression of osteogenic differentiation markers RUNX2 and OCN in MC3T3-E1 cells, and improved cell osteogenic capacity. However, these trends were significantly reversed in MC3T3-E1 cells following treatment with BMSCs-exo transfected with si-USP14. Furthermore, knockdown of USP14 promoted SIRT1 ubiquitination and degradation, the process that was reversed by the proteasome inhibitor MG132, whereas USP14 overexpression inhibited SIRT1 ubiquitination. In MC3T3-E1 cells infected with S. aureus, BMSCs-exo delivers USP14, which may enhance SIRT1 deubiquitination and increase SIRT1 protein activity. This process inhibits inflammation and promotes osteogenesis, warranting further investigation into its mechanisms and in vivo efficacy.
Collapse
Affiliation(s)
- Jun Yu
- Orthopaedic Trauma Department, General Hospital of Ningxia Medical University, 804 Shengli South Street, Xingqing District, Yinchuan City, Ningxia, 750001, China
| | - Ming Yang
- Emergency Department, People's Hospital of Ningxia Hui Autonomous Region, Ningxia, China
| | - Yun Jin
- Orthopaedic Trauma Department, General Hospital of Ningxia Medical University, 804 Shengli South Street, Xingqing District, Yinchuan City, Ningxia, 750001, China
| | - Kaijie Yang
- Orthopaedic Trauma Department, General Hospital of Ningxia Medical University, 804 Shengli South Street, Xingqing District, Yinchuan City, Ningxia, 750001, China
| | - Haibo Yang
- Orthopaedic Trauma Department, General Hospital of Ningxia Medical University, 804 Shengli South Street, Xingqing District, Yinchuan City, Ningxia, 750001, China.
| |
Collapse
|
4
|
Souza ATP, Freitas GP, Lopes HB, Weffort D, Adolpho LF, Gomes MPO, Oliveira FS, Almeida ALG, Beloti MM, Rosa AL. Diabetes Mellitus Impairs the Bone Regeneration Capacity of Mesenchymal Stromal Cell-Based Therapy. Arch Med Res 2025; 56:103234. [PMID: 40398343 DOI: 10.1016/j.arcmed.2025.103234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2025] [Revised: 03/15/2025] [Accepted: 04/23/2025] [Indexed: 05/23/2025]
Abstract
BACKGROUND Diabetes mellitus (DM) negatively impacts bone tissue, leading to bone loss and increased fracture risk. Many individuals need additional treatments, and therapy based on mesenchymal stromal cells (MSCs) represents a promising treatment for bone defects in patients with diabetes. AIMS The present study explored the effects of interactions between MSCs from normoglycemic (NG-MSCs) and diabetic (DM-MSCs) donors on osteoblast differentiation and the effects of cell therapy using NG-MSCs on bone regeneration in defects created in diabetic rats. METHODS After inducing DM with streptozotocin, we evaluated the morphometric parameters of rat femurs and the osteoblast differentiation of MSCs, as well as the effects of the interaction between NG-MSCs and DM-MSCs on their osteoblast differentiation. The efficacy of cell therapy was measured by evaluating the bone repair in calvarial defects of diabetic rats treated with local injections of either NG-MSCs or a vehicle. RESULTS DM induced bone loss and impaired the osteoblast differentiation of MSCs, which was partially restored by NG-MSCs, while the bone formation observed in defects treated with NG-MSCs and the vehicle was similar. CONCLUSION These results indicate that the beneficial effect of NG-MSCs on DM-MSCs did not translate into enhanced bone repair, mainly due to a hostile environment created by hyperglycemia, which compromised the ability of MSCs to induce bone formation.
Collapse
Affiliation(s)
- Alann Thaffarell Portilho Souza
- Bone Research Lab, Ribeirão Preto School of Dentistry, University of São Paulo, Ribeirão Preto, SP, Brazil; School of Dentistry, Metropolitan University Center of the Amazon, Belém, Brazil; Postgraduate Dentistry Program, CEUMA University, São Luís, Brazil
| | - Gileade Pereira Freitas
- Bone Research Lab, Ribeirão Preto School of Dentistry, University of São Paulo, Ribeirão Preto, SP, Brazil; School of Dentistry, Federal University of Goiás, Goiânia, Brazil
| | - Helena Bacha Lopes
- Bone Research Lab, Ribeirão Preto School of Dentistry, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Denise Weffort
- Bone Research Lab, Ribeirão Preto School of Dentistry, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Leticia Faustino Adolpho
- Bone Research Lab, Ribeirão Preto School of Dentistry, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Maria Paula Oliveira Gomes
- Bone Research Lab, Ribeirão Preto School of Dentistry, University of São Paulo, Ribeirão Preto, SP, Brazil
| | | | | | - Marcio Mateus Beloti
- Bone Research Lab, Ribeirão Preto School of Dentistry, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Adalberto Luiz Rosa
- Bone Research Lab, Ribeirão Preto School of Dentistry, University of São Paulo, Ribeirão Preto, SP, Brazil.
| |
Collapse
|
5
|
Yuan J, Li M, He X, Hou Q, Fu X, Hao J. A thermally stable bioactive chitosan scaffold with pH-responsive exosome adsorption and release function promotes wound healing. Int J Biol Macromol 2025; 306:141552. [PMID: 40024417 DOI: 10.1016/j.ijbiomac.2025.141552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 02/16/2025] [Accepted: 02/25/2025] [Indexed: 03/04/2025]
Abstract
Chitosan is an excellent carrier material for bioactive substances, and its binding ability is affected by the pH value of surrounding environments. Healthy skin is maintained in a slightly acidic environment, whereas the wound healing environment is normally neutral or slightly alkaline. In the present study, the authors proposed developing a thermally stable bioactive chitosan scaffold (T-CS) with pH-responsive exosome adsorption and release functions to promote wound healing. Our results revealed that T-CS could automatically capture exosomes from human umbilical cord mesenchymal stem cells in an acidic environment and release them in alkaline or neutral environments. The exosomes separated by T-CS and the traditional ultracentrifugation (UC) method exhibited similar size and protein markers. Furthermore, the exosomal biological activities of the T-CS (T-CS-E) and UC groups exhibited similar anti-inflammatory, proproliferation, promigration, and proendothelial cell-tube formation effects on human umbilical vein endothelial cells. Similar results were achieved in a mouse model by sustainably releasing exosomes. T-CS-E could facilitate wound healing by enhancing cell proliferation, inhibiting wound inflammation, and promoting vascularization. Therefore, this study developed a T-CS scaffold that integrates exosome isolation and application for wound healing, laying the foundation for future clinical use.
Collapse
Affiliation(s)
- Jifang Yuan
- Medical Innovation Research Department, PLA General Hospital and PLA Medical College, Beijing, 100853, China; PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration, Beijing 100048, China; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing 100048, China
| | - Meirong Li
- Center for Drug Evaluation, National Medical Products Administration, Beijing 100076, China
| | - Xiaofeng He
- Department of Diagnostic Radiology, The First Medical Center, Chinese PLA General Hospital, Beijing 100853, China
| | - Qian Hou
- Medical Innovation Research Department, PLA General Hospital and PLA Medical College, Beijing, 100853, China; PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration, Beijing 100048, China; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing 100048, China.
| | - Xiaobing Fu
- Medical Innovation Research Department, PLA General Hospital and PLA Medical College, Beijing, 100853, China; PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration, Beijing 100048, China; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing 100048, China.
| | - Jianxiu Hao
- Medical Innovation Research Department, PLA General Hospital and PLA Medical College, Beijing, 100853, China; PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration, Beijing 100048, China; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing 100048, China; Graduate school of the PLA General Hospital, Beijing 100853, China.
| |
Collapse
|
6
|
Zhang L, Ji C, Li Z, Jiwa H, Xie Z, Luo X, Luo J. Sonic Hedgehog potentiates BMP9-induced osteogenic differentiation of mesenchymal stem cells. Genes Dis 2025; 12:101308. [PMID: 40070367 PMCID: PMC11894376 DOI: 10.1016/j.gendis.2024.101308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Revised: 02/28/2024] [Accepted: 03/13/2024] [Indexed: 03/14/2025] Open
Abstract
Bone morphogenetic protein 9 (BMP9) has remarkable potential to induce the differentiation of mesenchymal stem cells (MSCs) towards the osteoblastic lineage. Additionally, research suggests that certain growth factors have the ability to potentiate BMP9-induced osteogenic differentiation of MSCs. Sonic Hedgehog (Shh) plays an indispensable role in the regulation of skeletal development. The objective of this research was to assess the potential influence of Shh on BMP9-induced osteogenic differentiation of MSCs. Our findings indicated that Shh effectively enhanced BMP9-induced early and late osteogenic differentiation of MSCs, and increased BMP9-induced expression/transcriptional activity of osteogenesis-related transcription factors. Besides, it was observed that Shh promoted BMP9-induced ectopic bone formation of MSCs in vivo. Moreover, BMP9 was able to facilitate the repair of bone defects in rats, while Shh further accelerated this reparative process. Mechanistically, Shh enhanced the activation of the Smad1/5/8 signaling pathway which was induced by BMP9. Furthermore, GANT-61, an inhibitor of Gli1 and Gli2, attenuated the enhancing effect of Shh on BMP9-induced osteogenic differentiation of MSCs. Collectively, the co-administration of BMP9 and Shh may present a promising therapeutic approach for the treatment of fracture nonunion, delayed fracture healing, and bone defects.
Collapse
Affiliation(s)
- Lulu Zhang
- Key Laboratory of Diagnostic Medicine Designated by the Chinese Ministry of Education, Chongqing Medical University, Chongqing 40016, China
| | - Caixia Ji
- Department of Clinical Laboratory, People's Hospital of Deyang City, Deyang, Sichuan 618000, China
| | - Ziyun Li
- Key Laboratory of Diagnostic Medicine Designated by the Chinese Ministry of Education, Chongqing Medical University, Chongqing 40016, China
| | - Habu Jiwa
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Zhou Xie
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Xiaoji Luo
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Jinyong Luo
- Key Laboratory of Diagnostic Medicine Designated by the Chinese Ministry of Education, Chongqing Medical University, Chongqing 40016, China
| |
Collapse
|
7
|
Borkowska P, Kowalczyk M, Zielińska A, Poskrobko K, Rother MB, Paul-Samojedny M, Kowalski J. NGF regulates survival and differentiation of umbilical mesenchymal stem/stromal cells into GABAergic, dopaminergic and cholinergic lineages. Eur J Pharm Sci 2025; 208:107053. [PMID: 40010415 DOI: 10.1016/j.ejps.2025.107053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 02/21/2025] [Accepted: 02/24/2025] [Indexed: 02/28/2025]
Abstract
Mesenchymal stem cells advantageous properties have led scientists to conduct trials on a range of medical conditions, including incurable neurodegenerative diseases. Wharton-Jelly derived mesenchymal stem cells, given their ease of collection, are frequently selected for these studies. This research aimed to investigate the effects of nerve growth factor (NGF) gene overexpression on the neural differentiation, survivability, and gene and protein expression of these cells. The level of gene expression was tested using the ddPCR method. Six umbilical cords from donors were collected, and three randomly chosen primary cultures of Wharton-Jelly derived mesenchymal stem cells were used in experiment. Cells were transduced with lentiviral vectors and underwent a 12-day differentiation process. The results revealed neuron-like cells with significantly high expression of CHAT, GAD2 and TH genes. A corresponding increase in protein expression was also observed. Immunostaining demonstrated notable differences in neuron-like phenotypes, contingent on the environmental conditions of the research groups. Throughout the experiment, samples with transduced mesenchymal stem cells overexpressing the NGF gene showed the highest expression levels from almost all of studied genes and proteins, and were also the most phenotypically similar to neuron-like cells. The study concluded that sustained overexpression of NGF: guides mesenchymal stem cells towards the neural pathway, facilitates the differentiation of modified mesenchymal stem cells into GABAergic, dopaminergic, and cholinergic neuron-like cells, suggests that GABAergic neurons' marker predominantly co-expresses with other neurons' markers, such as cholinergic or dopaminergic ones, increases survivability of modified mesenchymal stem cells in toxic conditions; The limitations of the study is that we merely know that cells have begun to express neurogenic markers, but in the absence of standards for mature neuronal markers, we do not yet know how far they have progressed as differentiating cells.
Collapse
Affiliation(s)
- Paulina Borkowska
- Department of Medical Genetics, Medical University of Silesia, Katowice, Poland.
| | - Małgorzata Kowalczyk
- Department of Medical Genetics, Medical University of Silesia, Katowice, Poland.
| | - Aleksandra Zielińska
- Department of Medical Genetics, Medical University of Silesia, Katowice, Poland.
| | - Karol Poskrobko
- Department of Medical Genetics, Medical University of Silesia, Katowice, Poland.
| | - Magdalena B Rother
- Department of Human Genetics, Leiden University Medical Center (LUMC), 2300 RC, The Netherlands
| | | | - Jan Kowalski
- Department of Medical Genetics, Medical University of Silesia, Katowice, Poland.
| |
Collapse
|
8
|
Haddadin L, Sun X. Stem Cells in Cancer: From Mechanisms to Therapeutic Strategies. Cells 2025; 14:538. [PMID: 40214491 PMCID: PMC11988674 DOI: 10.3390/cells14070538] [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: 03/02/2025] [Revised: 03/23/2025] [Accepted: 04/01/2025] [Indexed: 04/14/2025] Open
Abstract
Stem cells have emerged as a pivotal area of research in the field of oncology, offering new insights into the mechanisms of cancer initiation, progression, and resistance to therapy. This review provides a comprehensive overview of the role of stem cells in cancer, focusing on cancer stem cells (CSCs), their characteristics, and their implications for cancer therapy. We discuss the origin and identification of CSCs, their role in tumorigenesis, metastasis, and drug resistance, and the potential therapeutic strategies targeting CSCs. Additionally, we explore the use of normal stem cells in cancer therapy, focusing on their role in tissue regeneration and their use as delivery vehicles for anticancer agents. Finally, we highlight the challenges and future directions in stem cell research in cancer.
Collapse
Affiliation(s)
| | - Xueqin Sun
- Cancer Genome and Epigenetics Program, NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| |
Collapse
|
9
|
Goulian AJ, Goldstein B, Saad MA. Advancements in Regenerative Therapies for Orthopedics: A Comprehensive Review of Platelet-Rich Plasma, Mesenchymal Stem Cells, Peptide Therapies, and Biomimetic Applications. J Clin Med 2025; 14:2061. [PMID: 40142869 PMCID: PMC11943164 DOI: 10.3390/jcm14062061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Revised: 03/09/2025] [Accepted: 03/12/2025] [Indexed: 03/28/2025] Open
Abstract
Background/Objectives: Regenerative therapies have gained interest in orthopedic applications for their potential to enhance tissue regeneration, functional recovery, and pain modification. This review evaluates the clinical efficacy of platelet-rich plasma (PRP), mesenchymal stem cells (MSCs), peptide-based treatments, and biomimetic materials in orthopedic care, with a focus on pain reduction and functional outcomes. Methods: A structured literature search in PubMed (January 2009-January 2025) identified 160 studies. After applying inclusion criteria prioritizing randomized controlled trials (RCTs) and clinical trials, 59 studies were included: 20 on PRP, 20 on MSCs, 10 on peptide therapies, and 7 on biomimetics. Data extraction focused on pain reduction and functional recovery, with risk of bias assessed using the Cochrane Risk of Bias (RoB) tool and ROBINS-I tool. A random-effects meta-regression analysis was conducted to evaluate the impact of therapy type, sample size, and risk of bias on reported pain reduction outcomes. Results: Meta-regression analysis identified MSC therapy as the most effective intervention for pain reduction (β = 8.45, p < 0.05), with PRP and peptide-based therapies showing moderate improvements, and biomimetic therapies demonstrating the lowest effect. PRP provided short-term pain relief, particularly in acute injuries and tendon repair, though inconsistencies in preparation methods limited success in chronic conditions. MSC therapies demonstrated cartilage regeneration and early osteoarthritis improvement, but high costs and ethical concerns remain barriers to widespread adoption. Peptide-based therapies and biomimetic materials, including engineered scaffolds and autologous protein solutions, showed promise for infection control and wound healing, though further research is needed to optimize dosing, delivery methods, and long-term safety. Conclusions: Regenerative therapies offer significant potential in orthopedic care, with MSC therapies demonstrating the most reliable regenerative effects, PRP providing short-term symptomatic relief, and peptide-based and biomimetic treatments emerging as promising adjuncts. However, standardized protocols and large-scale clinical trials are needed to establish long-term efficacy and improve clinical translation for broader adoption.
Collapse
Affiliation(s)
- Andrew J. Goulian
- College of Medicine, California Northstate University, Elk Grove, CA 95757, USA; (A.J.G.); (B.G.)
| | - Brielle Goldstein
- College of Medicine, California Northstate University, Elk Grove, CA 95757, USA; (A.J.G.); (B.G.)
| | - Maarouf A. Saad
- Department of Orthopaedic Surgery, University of California, Sacramento, CA 95817, USA
| |
Collapse
|
10
|
Shen S, Shu B, Xu Y, Zhao H, Li Y, Li Y, Zhuo C, Zhuo N. Characterization and Biocompatibility Assessment of 3D-Printed HA/PCL Porous Bionic Bone Scaffold: in Vitro and in Vivo Evaluation. JOURNAL OF MUSCULOSKELETAL & NEURONAL INTERACTIONS 2025; 25:119-132. [PMID: 40024235 PMCID: PMC11880846 DOI: 10.22540/jmni-25-119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Accepted: 11/21/2024] [Indexed: 03/04/2025]
Abstract
OBJECTIVES This study aims to characterize a three-dimensional-printed hydroxyapatite (HA)/polycaprolactone (PCL) scaffold and assess its biocompatibility both in vitro and in vivo. METHODS A bionic, porous HA/PCL scaffold was fabricated using 3D printing, and its microstructure, porosity, hydrophilicity, and mechanical properties were evaluated through scanning electron microscopy and various assays. Bone marrow mesenchymal stem cells (BMSCs) and vascular endothelial progenitor cells (VEPCs) were co-cultured with the scaffold, and their proliferation and osteogenic differentiation were assessed using the Cell Counting Kit-8, ALP assays, and alizarin red staining. Osteogenic marker expression was analyzed via qRT-PCR. In vivo bone regeneration was evaluated through histological analysis of H&E and Masson's trichrome staining in a rat cranial defect model. RESULTS The average pore size of the scaffold was 462.00 ± 100.389 μm, with a porosity of 53%, a water absorption expansion rate of 5.10%, a contact angle of 94.55°, an elastic modulus of 53.82 MPa, and a compressive strength of 6.10 MPa. ALP activity and qRT-PCR analysis of osteogenic markers (BMP2, OCN, Runx2) showed significant upregulation in cells co-cultured with the scaffolds. In vivo experiments demonstrated enhanced bone regeneration and collagen deposition in the HA/PCL scaffold group. CONCLUSION The results suggest that the HA/PCL scaffold promotes osteogenic differentiation and bone regeneration, making it suitable for bone tissue engineering applications.
Collapse
Affiliation(s)
- Shi Shen
- Department of Bone and Joint Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Benchao Shu
- Department of Bone and Joint Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Department of Joint Trauma Surgery, Guangyuan First People’s Hospital, Guangyuan, Sichuan, China
| | - Yulin Xu
- Department of Bone and Joint Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Heng Zhao
- Department of Bone and Joint Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Yang Li
- Department of Bone and Joint Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Yujie Li
- Department of Bone and Joint Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Chuanchuan Zhuo
- Department of Bone and Joint Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Naiqiang Zhuo
- Department of Bone and Joint Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| |
Collapse
|
11
|
Das S, Thakur A, Datta A, Sahoo A, Bandyopadhyay S, Sah AK. Advances in Regenerative Medicine for Orthopedic Injuries: A Comprehensive Review. Cureus 2025; 17:e79860. [PMID: 40166527 PMCID: PMC11956119 DOI: 10.7759/cureus.79860] [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] [Accepted: 02/28/2025] [Indexed: 04/02/2025] Open
Abstract
Orthopedics is one field that greatly benefits from the new ideas provided by regenerative medicine. This review pulls together the most recent publications involving stem cell therapy, platelet-rich plasma, growth factor, gene therapy, tissue engineering, stem cell-derived extracellular vesicles, and other regenerative technologies in the context of bone, cartilage, tendon, and ligament healing. Recent studies show that these new therapies can alter cell development, division, and production of fiber and ground substance to remodel tissues. Nevertheless, the clinical application has several issues such as the standardization of cell procurement and preparation, the control of cytokine/gene delivery, the revascularization of tissues, and the requirements of large samples, positively controlled clinical trials. More research must be conducted to overcome such barriers and make practicing more applicable in real life.
Collapse
Affiliation(s)
- Samyabrata Das
- Orthopaedic Surgery, Sri Ramachandra Institute of Higher Education and Research, Chennai, IND
| | - Amit Thakur
- Department of Orthopaedics and Traumatology, All India Institute of Medical Science Jammu, Jammu, IND
| | - Anupam Datta
- Forensic Medicine, Agartala Government Medical College & Govind Ballabh Pant (GBP) Hospital, Agartala, IND
| | - Ayaskant Sahoo
- Anaesthesia, NRI Institute of Medical Sciences, Visakhapatnam, IND
| | | | - Ashok K Sah
- Department of Medical Laboratory Sciences, College of Applied and Health Sciences, A Sharqiyah University, Ibra, OMN
| |
Collapse
|
12
|
Jia J, Wang L, Zhou Y, Zhang P, Chen X. Muscle-derived extracellular vesicles mediate crosstalk between skeletal muscle and other organs. Front Physiol 2025; 15:1501957. [PMID: 39844898 PMCID: PMC11750798 DOI: 10.3389/fphys.2024.1501957] [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: 09/26/2024] [Accepted: 12/18/2024] [Indexed: 01/24/2025] Open
Abstract
Skeletal muscle (SKM) has crucial roles in locomotor activity and posture within the body and also functions have been recognized as an actively secretory organ. Numerous bioactive molecules are secreted by SKM and transported by extracellular vesicles (EVs), a novel class of mediators of communication between cells and organs that contain various types of cargo molecules including lipids, proteins and nucleic acids. SKM-derived EVs (SKM-EVs) are intercellular communicators with significant roles in the crosstalk between SKM and other organs. In this review, we briefly describe the biological characteristics, composition, and uptake mechanisms of EVs, particularly exosomes, comprehensively summarize the regulatory effects of SKM-EVs on the function of, which include myogenesis, muscle repair and regeneration, as well as metabolic regulation. Furthermore, we explore the impact of SKM- EVs on various organs including bone, the cardiovascular system, adipose tissue, and nervous system. As emerging evidence suggests that SKM-EVs are involved in the development and regulation of type 2 diabetes (T2D), systemic inflammation, and other chronic diseases, we also highlight the potential of SKM-EVs as therapeutic targets and diagnostic biomarkers, emphasizing the need for further research to elucidate the complex mechanisms underlying intercellular communication in physiological and pathological contexts.
Collapse
Affiliation(s)
- Jiajie Jia
- National Key Laboratory of Human Factors Engineering, China Astronaut Research and Training Center, Beijing, China
- Department of Exercise Physiology, Beijing Sport University, Beijing, China
| | - Lu Wang
- National Key Laboratory of Human Factors Engineering, China Astronaut Research and Training Center, Beijing, China
- Department of Exercise Physiology, Beijing Sport University, Beijing, China
| | - Yue Zhou
- Department of Exercise Physiology, Beijing Sport University, Beijing, China
| | - Peng Zhang
- National Key Laboratory of Space Medicine, China Astronaut Research and Training Center, Beijing, China
| | - Xiaoping Chen
- National Key Laboratory of Human Factors Engineering, China Astronaut Research and Training Center, Beijing, China
| |
Collapse
|
13
|
Han D, Wang W, Gong J, Ma Y, Li Y. Controlled delivery of mesenchymal stem cells via biodegradable scaffolds for fracture healing. Nanomedicine (Lond) 2025; 20:207-224. [PMID: 39686770 PMCID: PMC11731254 DOI: 10.1080/17435889.2024.2439242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 12/04/2024] [Indexed: 12/18/2024] Open
Abstract
Biodegradable controlled delivery systems for mesenchymal stem cells (MSCs) have emerged as novel advancements in the field of regenerative medicine, particularly for accelerating bone fracture healing. This detailed study emphasizes the importance of quick and adequate fracture treatment and the limitations of existing methods. New approaches employing biodegradable scaffolds can be placed within a fracture to serve as a mechanical support and allow controlled release of in situ MSCs and bioactive agents. They are made up of polymers and composites which degrade over time, aiding in natural tissue regrowth. The fabrication methods, including 3D printing, electrospinning, and solvent casting, with particulate leaching that enable precise control over scaffold architecture and properties, are discussed. Progress in controlled drug delivery systems including encapsulation techniques and release kinetics is described, highlighting the potential of such strategies to maintain therapeutic benefits over a prolonged time as well as improving outcomes for fracture repair. MSCs play a role in bone regeneration through differentiation using biodegradable scaffolds, paracrine effects, and regulation of inflammation focusing on fracture healing. Current trends and future directions in scaffold technology and MSC delivery, including smart scaffolds with growth factor incorporation and innovative delivery approaches for fracture healing are also discussed.
Collapse
Affiliation(s)
- Dong Han
- Trauma Orthopedics Department, Yantaishan Hospital, Yantai, China
| | - Weijiao Wang
- Otolaryngology Department, Yantaishan Hospital, Yantai, China
| | - Jinpeng Gong
- Trauma Orthopedics Department, Yantaishan Hospital, Yantai, China
| | - Yupeng Ma
- Trauma Orthopedics Department, Yantaishan Hospital, Yantai, China
| | - Yu Li
- Trauma Orthopedics Department, Yantaishan Hospital, Yantai, China
| |
Collapse
|
14
|
Yang J, Xiao L, Zhang L, Luo G, Ma Y, Wang X, Zhang Y. Platelets: A Potential Factor that Offers Strategies for Promoting Bone Regeneration. TISSUE ENGINEERING. PART B, REVIEWS 2024; 30:631-643. [PMID: 38482796 DOI: 10.1089/ten.teb.2024.0004] [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: 04/13/2024]
Abstract
Bone defects represent a prevalent category of clinical injuries, causing significant pain and escalating health care burdens. Effectively addressing bone defects is thus of paramount importance. Platelets, formed from megakaryocyte lysis, have emerged as pivotal players in bone tissue repair, inflammatory responses, and angiogenesis. Their intracellular storage of various growth factors, cytokines, and membrane protein receptors contributes to these crucial functions. This article provides a comprehensive overview of platelets' roles in hematoma structure, inflammatory responses, and angiogenesis throughout the process of fracture healing. Beyond their application in conjunction with artificial bone substitute materials for treating bone defects, we propose the potential future use of anticoagulants such as heparin in combination with these materials to regulate platelet number and function, thereby promoting bone healing. Ultimately, we contemplate whether manipulating platelet function to modulate bone healing could offer innovative ideas and directions for the clinical treatment of bone defects. Impact statement Given that 5-10% of fracture patients with delayed bone healing or even bone nonunion, this review explores the potential role of platelets in bone healing (directly/indirectly) and proposes ideas and directions for the future as to whether it is possible to promote bone healing and improve fracture healing rates by modulating platelets.
Collapse
Affiliation(s)
- Jingjing Yang
- Department of Hygiene Toxicology, School of Public Health, Zunyi Medical University, Zunyi, China
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China
- Key Laboratory of Maternal and Child Health and Exposure Science of Guizhou Higher Education Institutes, Zunyi Medical University, Zunyi, China
- Guizhou Provincial Key Laboratory of Medicinal Biotechnology in Colleges and Universities, Zunyi Medical University, Zunyi, China
| | - Lan Xiao
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China
- School of Medicine and Dentistry, Griffith University, Queensland, Australia
| | - Lijia Zhang
- Department of Hygiene Toxicology, School of Public Health, Zunyi Medical University, Zunyi, China
- Key Laboratory of Maternal and Child Health and Exposure Science of Guizhou Higher Education Institutes, Zunyi Medical University, Zunyi, China
| | - Guochen Luo
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Yaping Ma
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Xin Wang
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China
- Guizhou Provincial Key Laboratory of Medicinal Biotechnology in Colleges and Universities, Zunyi Medical University, Zunyi, China
| | - Yi Zhang
- Department of Hygiene Toxicology, School of Public Health, Zunyi Medical University, Zunyi, China
- Key Laboratory of Maternal and Child Health and Exposure Science of Guizhou Higher Education Institutes, Zunyi Medical University, Zunyi, China
| |
Collapse
|
15
|
Tsai SC, Wan BH, Tsai FJ, Yang JS. Artificial intelligence (AI)-powered bibliometric analysis of global trends in mesenchymal stem cells (MSCs)-derived exosome research: 2014-2023. Biomedicine (Taipei) 2024; 14:61-77. [PMID: 39777112 PMCID: PMC11703396 DOI: 10.37796/2211-8039.1470] [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: 09/30/2024] [Accepted: 10/21/2024] [Indexed: 01/11/2025] Open
Abstract
Introduction In recent years, significant progress has been made in regenerative medicine, specifically in using mesenchymal stem cells (MSCs) due to their regenerative and differentiating abilities. An exciting development in this area is the utilization of exosomes derived from MSCs, which have shown promise in tissue restoration, immune system modulation, and cancer treatment. Objectives This study aims to analyze global research trends and the academic impact of MSCs-derived exosomes from 2014 to 2023, providing a comprehensive overview of this emerging field. Materials and methods The Web of Science database selected 948 relevant publications from 2014 to 2023. Artificial intelligence (AI)-bibliometric tools, including Bibliometrix, CiteSpace, and VOSviewer, were employed to analyze and visualize the data. The focus was on publication quantity, research nations, institutional partnerships, keywords, and research focal points. Results The study revealed that China, Japan, Taiwan, and the United States are the leaders in publication volume and impact in MSCs-derived exosome research. China has the highest number of publications, while the United States and Iran excel in research quality and influence. Primary research themes were identified through keyword and clustering analyses, including tissue repair, immune modulation, bone regeneration, and cancer treatment. The study also emphasized the importance of international collaboration, with China and the United States demonstrating the most robust cooperation. Conclusion MSCs-derived exosome research rapidly expands worldwide, showing promising prospects in regenerative medicine and cell therapy. With continued research and international collaboration, MSCs-derived exosomes are expected to play a vital role in future therapeutic application.
Collapse
Affiliation(s)
- Shih-Chang Tsai
- Department of Biological Science and Technology, China Medical University, Taichung,
Taiwan
| | - Bing-Han Wan
- Department of Biological Science and Technology, China Medical University, Taichung,
Taiwan
| | - Fuu-Jen Tsai
- School of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung,
Taiwan
- China Medical University Children’s Hospital, Taichung,
Taiwan
- Department of Medical Genetics, China Medical University Hospital, Taichung,
Taiwan
| | - Jai-Sing Yang
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung,
Taiwan
| |
Collapse
|
16
|
Gupta S, Qayoom I, Mairal A, Singh S, Kumar A. Local Delivery of Exosomes and Antibiotics in Hydroxyapatite-Based Nanocement for Osteomyelitis Management. ACS Infect Dis 2024; 10:3994-4008. [PMID: 39469832 DOI: 10.1021/acsinfecdis.4c00721] [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: 10/30/2024]
Abstract
The management of bone and joint infections is a formidable challenge in orthopedics and poses a global health concern. While clinical management emphasizes infection prevention and complete eradication, an effective strategy for stabilizing skeletal tissue with adequate soft tissue coverage remains limited. In this study, we have employed a novel approach of using the local delivery of exosomes and antibiotics (rifampicin) using a hydroxyapatite-based nanocement carrier to manage the residual space created during debridement effectively. Additionally, we synthesized a periosteum-guiding antioxidant herbal membrane to leverage the inherent periosteum regeneration capability of the bone, facilitating bone callus repair and natural healing. The synthesized scaffolds were biocompatible and demonstrated potent antibacterial activity in vitro. When evaluated in vivo in the Staphylococcus aureus-induced rat tibial osteomyelitis model, the released drugs successfully cleared the residual bacteria and the released exosome promoted bone healing, resulting in 3-fold increase in bone volume as analyzed via micro-CT analysis. Immunofluorescence staining of periosteum-specific markers also indicated the complete formation of periosteal layers, thus highlighting the complete bone repair.
Collapse
Affiliation(s)
- Sneha Gupta
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, UP 208016, India
| | - Irfan Qayoom
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, UP 208016, India
| | - Ayushi Mairal
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, UP 208016, India
| | - Sneha Singh
- Department of Bioengineering and Biotechnology, Birla Institute of Technology, Mesra, Ranchi, Jharkhand 835215, India
| | - Ashok Kumar
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, UP 208016, India
- Centre for Environmental Science and Engineering, Indian Institute of Technology Kanpur, Kanpur, UP 208016, India
- The Mehta Family Centre for Engineering in Medicine, Indian Institute of Technology Kanpur, Kanpur, UP 208016, India
- Centre for Nanosciences, Indian Institute of Technology Kanpur, Kanpur, UP 208016, India
- Centre of Excellence in Orthopaedics and Prosthetics, Gangwal School of Medical Sciences and Technology, Indian Institute of Technology Kanpur, Kanpur, UP 208016, India
| |
Collapse
|
17
|
Zhang Y, Fan M, Zhang Y. Revolutionizing bone defect healing: the power of mesenchymal stem cells as seeds. Front Bioeng Biotechnol 2024; 12:1421674. [PMID: 39497791 PMCID: PMC11532096 DOI: 10.3389/fbioe.2024.1421674] [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: 04/22/2024] [Accepted: 10/10/2024] [Indexed: 11/07/2024] Open
Abstract
Bone defects can arise from trauma or pathological factors, resulting in compromised bone integrity and the loss or absence of bone tissue. As we are all aware, repairing bone defects is a core problem in bone tissue engineering. While minor bone defects can self-repair if the periosteum remains intact and normal osteogenesis occurs, significant defects or conditions such as congenital osteogenesis imperfecta present substantial challenges to self-healing. As research on mesenchymal stem cell (MSC) advances, new fields of application have emerged; however, their application in orthopedics remains one of the most established and clinically valuable directions. This review aims to provide a comprehensive overview of the research progress regarding MSCs in the treatment of diverse bone defects. MSCs, as multipotent stem cells, offer significant advantages due to their immunomodulatory properties and ability to undergo osteogenic differentiation. The review will encompass the characteristics of MSCs within the osteogenic microenvironment and summarize the research progress of MSCs in different types of bone defects, ranging from their fundamental characteristics and animal studies to clinical applications.
Collapse
Affiliation(s)
- Yueyao Zhang
- Trauma Emergency Center, The Third Hospital of Hebei Medical University, Shijiazhuang, China
- Key Laboratory of Biomechanics of Hebei Province, Orthopaedic Research Institution of Hebei Province, Shijiazhuang, China
| | - Mengke Fan
- Trauma Emergency Center, The Third Hospital of Hebei Medical University, Shijiazhuang, China
- Key Laboratory of Biomechanics of Hebei Province, Orthopaedic Research Institution of Hebei Province, Shijiazhuang, China
| | - Yingze Zhang
- Trauma Emergency Center, The Third Hospital of Hebei Medical University, Shijiazhuang, China
- Key Laboratory of Biomechanics of Hebei Province, Orthopaedic Research Institution of Hebei Province, Shijiazhuang, China
| |
Collapse
|
18
|
Guo J, Shu X, Yu S, Guo C, Shen G, Chen L, Zhou J, Xiao J, Guo H, Chen Y, Zeng Z, Wang P. Injectable hydrogel microsphere-bomb for MRSA-infected chronic osteomyelitis. J Control Release 2024; 376:337-353. [PMID: 39413850 DOI: 10.1016/j.jconrel.2024.10.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 10/08/2024] [Accepted: 10/13/2024] [Indexed: 10/18/2024]
Abstract
Biofilm and bone tissue defect induced by the bacterial infection severely impede chronic osteomyelitis treatment. It is critical to break though the densely and obstinate biofilm so that the target drugs can deliver to the infected bone more effectively. Herein, an acoustically responsive multifunctional hydrogel microsphere-bomb (EMgel) was designed and prepared by microfluidic technology, which could be injected to the focus of bone infection, and blasted into the nidus deeply to destroy the bacterial biofilm matrix barrier under penetrating ultrasound, so the encapsulated natural polyphenolic EGCG and bioactive MoS2 released to repair the damaged bone. The results proved the hydrogel microsphere-bomb exhibited controlling drug release, favorable antibacterial (as high as 99 %), high biofilm resistance, fascinating antioxidation, good cytocompatibility, and osteogenic differentiation. The acoustically responsive microsphere-bomb further proved their fantastic ability to eradicate biofilm and promote bone regeneration in the Methicillin-resistant Staphylococcus aureus (MRSA) infected chronic osteomyelitis model due to the synergy effects of EGCG and bioactive MoS2. Especially, immunohistochemical staining showed lower inflammatory reaction and higher expression of OCN in EMgel group treated with ultrasound wave. This study presents a new design of hydrogel microsphere-based intelligence drug delivery for osteomyelitis treatment, which exhibit great promising potential for dealing with chronic orthopedic infections, drug delivery system and tissue engineering.
Collapse
Affiliation(s)
- Jiayi Guo
- Department of Ultrasound, The Third Affiliated Hospital, Southern Medical University, Guangzhou 510630, China; Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou 510316, China
| | - Xian Shu
- Department of Ultrasound, The Third Affiliated Hospital, Southern Medical University, Guangzhou 510630, China
| | - Shan Yu
- Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou 510316, China
| | - Cuiping Guo
- Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou 510316, China
| | - Guangxin Shen
- Foshan Clinical Medical School of Guangzhou University of Chinese Medicine, Guangdong Province, Foshan 528031, China
| | - Longsheng Chen
- Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou 510316, China
| | - Jiayi Zhou
- Department of Ultrasound, The Third Affiliated Hospital, Southern Medical University, Guangzhou 510630, China
| | - Jiangwei Xiao
- Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou 510316, China
| | - Huilong Guo
- Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou 510316, China
| | - Yi Chen
- Department of Ultrasound, The Third Affiliated Hospital, Southern Medical University, Guangzhou 510630, China
| | - Zhiwen Zeng
- Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou 510316, China; Guangdong Key Lab of Medical Electronic Instruments and Polymer Material Products, Guangzhou 510500, China; National Engineering Research Center for Healthcare Devices, Guangzhou 510500, China.
| | - Ping Wang
- Department of Ultrasound, The Third Affiliated Hospital, Southern Medical University, Guangzhou 510630, China.
| |
Collapse
|
19
|
Trapana J, Weinerman J, Lee D, Sedani A, Constantinescu D, Best TM, Hornicek FJ, Hare JM. Cell-based therapy in the treatment of musculoskeletal diseases. Stem Cells Transl Med 2024; 13:959-978. [PMID: 39226104 PMCID: PMC11465182 DOI: 10.1093/stcltm/szae049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 05/21/2024] [Indexed: 09/04/2024] Open
Abstract
A limited number of tissues can spontaneously regenerate following injury, and even fewer can regenerate to a state comparable to mature, healthy adult tissue. Mesenchymal stem cells (MSCs) were first described in the 1960s-1970s by Friedenstein et al as a small population of bone marrow cells with osteogenic potential and abilities to differentiate into chondrocytes. In 1991, Arnold Caplan coined the term "mesenchymal cells" after identifying these cells as a theoretical precursor to bone, cartilage, tendon, ligament, marrow stroma, adipocyte, dermis, muscle, and connective tissues. MSCs are derived from periosteum, fat, and muscle. Another attractive property of MSCs is their immunoregulatory and regenerative properties, which result from crosstalk with their microenvironment and components of the innate immune system. Collectively, these properties make MSCs potentially attractive for various therapeutic purposes. MSCs offer potential in sports medicine, aiding in muscle recovery, meniscal tears, and tendon and ligament injuries. In joint disease, MSCs have the potential for chondrogenesis and reversing the effects of osteoarthritis. MSCs have also demonstrated potential application to the treatment of degenerative disc disease of the cervical, thoracic, and lumbar spine.
Collapse
Affiliation(s)
- Justin Trapana
- Department of Orthopaedics, University of Miami Miller School of Medicine, Miami, United States
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, United States
| | - Jonathan Weinerman
- Department of Orthopaedics, University of Miami Miller School of Medicine, Miami, United States
| | - Danny Lee
- Department of Orthopaedics, University of Miami Miller School of Medicine, Miami, United States
| | - Anil Sedani
- Department of Orthopaedics, University of Miami Miller School of Medicine, Miami, United States
| | - David Constantinescu
- Department of Orthopaedics, University of Miami Miller School of Medicine, Miami, United States
| | - Thomas M Best
- Department of Orthopaedics, University of Miami Miller School of Medicine, Miami, United States
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, United States
| | - Francis J Hornicek
- Department of Orthopaedics, University of Miami Miller School of Medicine, Miami, United States
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, United States
| | - Joshua M Hare
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, United States
| |
Collapse
|
20
|
Chen H, Wu X, Lan Y, Zhou X, Zhang Y, Long L, Zhong Y, Hao Z, Zhang W, Xue D. SCUBE3 promotes osteogenic differentiation and mitophagy in human bone marrow mesenchymal stem cells through the BMP2/TGF-β signaling pathway. FASEB J 2024; 38:e70011. [PMID: 39250278 DOI: 10.1096/fj.202400991r] [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: 05/02/2024] [Revised: 07/21/2024] [Accepted: 08/12/2024] [Indexed: 09/11/2024]
Abstract
In clinical settings, addressing large bone defects remains a significant challenge for orthopedic surgeons. The use of genetically modified bone marrow mesenchymal stem cells (BMSCs) has emerged as a highly promising approach for these treatments. Signal peptide-CUB-EGF domain-containing protein 3 (SCUBE3) is a multifunctional secreted glycoprotein, the role of which remains unclear in human hBMSCs. This study used various experimental methods to elucidate the potential mechanism by which SCUBE3 influences osteogenic differentiation of hBMSCs in vitro. Additionally, the therapeutic efficacy of SCUBE3, in conjunction with porous GeLMA microspheres, was evaluated in vivo using a mouse bone defect model. Our findings indicate that SCUBE3 levels increase significantly during early osteogenic differentiation of hBMSCs, and that reducing SCUBE3 levels can hinder this differentiation. Overexpressing SCUBE3 elevated osteogenesis gene and protein levels and enhanced calcium deposition. Furthermore, treatment with recombinant human SCUBE3 (rhSCUBE3) protein boosted BMP2 and TGF-β expression, activated mitophagy in hBMSCs, ameliorated oxidative stress, and restored osteogenic function through SMAD phosphorylation. In vivo, GELMA/OE treatment effectively accelerated bone healing in mice. In conclusion, SCUBE3 fosters osteogenic differentiation and mitophagy in hBMSCs by activating the BMP2/TGF-β signaling pathway. When combined with engineered hydrogel cell therapy, it could offer valuable guidance for the clinical management of extensive bone defects.
Collapse
Affiliation(s)
- Hongyu Chen
- Department of Orthopaedics, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, P.R. China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, P.R. China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, P.R. China
| | - Xiaoyong Wu
- Department of Orthopaedics, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, P.R. China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, P.R. China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, P.R. China
| | - Yinan Lan
- Department of Orthopaedics, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, P.R. China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, P.R. China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, P.R. China
| | - Xijie Zhou
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Ye Zhang
- Department of Orthopaedics, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, P.R. China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, P.R. China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, P.R. China
| | - Long Long
- Department of Orthopaedics, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, P.R. China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, P.R. China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, P.R. China
| | - Yuliang Zhong
- Department of Orthopaedics, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, P.R. China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, P.R. China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, P.R. China
| | - Zhengan Hao
- Department of Orthopaedics, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, P.R. China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, P.R. China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, P.R. China
| | - Weijun Zhang
- Department of Orthopaedics, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, P.R. China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, P.R. China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, P.R. China
| | - DeTing Xue
- Department of Orthopaedics, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, P.R. China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, P.R. China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, P.R. China
| |
Collapse
|
21
|
Yao Q, He L, Bao C, Yan X, Ao J. The role of TNF-α in osteoporosis, bone repair and inflammatory bone diseases: A review. Tissue Cell 2024; 89:102422. [PMID: 39003912 DOI: 10.1016/j.tice.2024.102422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2023] [Revised: 05/17/2024] [Accepted: 05/23/2024] [Indexed: 07/16/2024]
Abstract
Tumour necrosis factor alpha (TNF-α) is a pleiotropic cytokine synthesised primarily by mononuclear cells; it has a potent pro-inflammatory effect, playing a crucial role in metabolic, immune, and inflammatory diseases. This cytokine has been studied in various biological systems. In bone tissue, TNF-α plays an integral role in skeletal disorders such as osteoporosis, fracture repair and rheumatoid arthritis through its involvement in regulating the balance between osteoblasts and osteoclasts, mediating inflammatory responses, promoting angiogenesis and exacerbating synovial proliferation. The biological effect TNF-α exerts in this context is determined by a combination of the signalling pathway it activates, the type of receptor it binds, and the concentration and duration of exposure. This review summarises the participation and pathophysiological role of TNF-α in osteoporosis, bone damage repair, chronic immunoinflammatory bone disease and spinal cord injury, and discusses its main mechanisms.
Collapse
Affiliation(s)
| | - Li He
- Affiliated Hospital of Zunyi Medical University, China.
| | | | - Xuhang Yan
- Affiliated Hospital of Zunyi Medical University, China.
| | - Jun Ao
- Affiliated Hospital of Zunyi Medical University, China.
| |
Collapse
|
22
|
Ji Y, Mao Y, Lin H, Wang Y, Zhao P, Guo Y, Gu L, Fu C, Chen X, Lv Z, Wang N, Li Q, Bei C. Acceleration of bone repairation by BMSCs overexpressing NGF combined with NSA and allograft bone scaffolds. Stem Cell Res Ther 2024; 15:194. [PMID: 38956719 PMCID: PMC11218317 DOI: 10.1186/s13287-024-03807-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Accepted: 06/18/2024] [Indexed: 07/04/2024] Open
Abstract
BACKGROUND Repairation of bone defects remains a major clinical problem. Constructing bone tissue engineering containing growth factors, stem cells, and material scaffolds to repair bone defects has recently become a hot research topic. Nerve growth factor (NGF) can promote osteogenesis of bone marrow mesenchymal stem cells (BMSCs), but the low survival rate of the BMSCs during transplantation remains an unresolved issue. In this study, we investigated the therapeutic effect of BMSCs overexpression of NGF on bone defect by inhibiting pyroptosis. METHODS The relationship between the low survival rate and pyroptosis of BMSCs overexpressing NGF in localized inflammation of fractures was explored by detecting pyroptosis protein levels. Then, the NGF+/BMSCs-NSA-Sca bone tissue engineering was constructed by seeding BMSCs overexpressing NGF on the allograft bone scaffold and adding the pyroptosis inhibitor necrosulfonamide(NSA). The femoral condylar defect model in the Sprague-Dawley (SD) rat was studied by micro-CT, histological, WB and PCR analyses in vitro and in vivo to evaluate the regenerative effect of bone repair. RESULTS The pyroptosis that occurs in BMSCs overexpressing NGF is associated with the nerve growth factor receptor (P75NTR) during osteogenic differentiation. Furthermore, NSA can block pyroptosis in BMSCs overexpression NGF. Notably, the analyses using the critical-size femoral condylar defect model indicated that the NGF+/BMSCs-NSA-Sca group inhibited pyroptosis significantly and had higher osteogenesis in defects. CONCLUSION NGF+/BMSCs-NSA had strong osteogenic properties in repairing bone defects. Moreover, NGF+/BMSCs-NSA-Sca mixture developed in this study opens new horizons for developing novel tissue engineering constructs.
Collapse
Affiliation(s)
- Ying Ji
- Department of Orthopaedics, Affiliated Hospital of Guilin Medical University, 15 Lequn Road, Guilin, 541001, China
| | - Yongkang Mao
- Department of Orthopaedics, Affiliated Hospital of Guilin Medical University, 15 Lequn Road, Guilin, 541001, China
| | - Honghu Lin
- Department of Orthopaedics, Affiliated Hospital of Guilin Medical University, 15 Lequn Road, Guilin, 541001, China
| | - Ye Wang
- Department of Orthopaedics, Affiliated Hospital of Guilin Medical University, 15 Lequn Road, Guilin, 541001, China
| | - Peishuai Zhao
- Department of Orthopaedics, Affiliated Hospital of Guilin Medical University, 15 Lequn Road, Guilin, 541001, China
| | - Yong Guo
- Department of Biomedical Engineering, School of Intelligent Medicine and Biotechnology, Guilin Medical University, 1 Zhiyuan Road, Guilin, 541199, China
| | - Lantao Gu
- Key Laboratory of Medical Biotechnology and Translational Medicine, Guilin Medical University, 1 Zhiyuan Road, Guilin, 541199, China
| | - Can Fu
- Key Laboratory of Medical Biotechnology and Translational Medicine, Guilin Medical University, 1 Zhiyuan Road, Guilin, 541199, China
| | - Ximiao Chen
- Department of Orthopaedics, Affiliated Hospital of Guilin Medical University, 15 Lequn Road, Guilin, 541001, China
| | - Zheng Lv
- Department of Radiology, Affiliated Hospital of Guilin Medical University, 15 Lequn Road, Guilin, 541001, China
| | - Ning Wang
- Department of Orthopaedics, Affiliated Hospital of Guilin Medical University, 15 Lequn Road, Guilin, 541001, China
| | - Qiang Li
- Department of Orthopaedics, Affiliated Hospital of Guilin Medical University, 15 Lequn Road, Guilin, 541001, China.
| | - Chaoyong Bei
- Department of Orthopaedics, Affiliated Hospital of Guilin Medical University, 15 Lequn Road, Guilin, 541001, China.
| |
Collapse
|
23
|
Altememy D, Kashani MHG, Fateme A, Khosravian P. New method to induce neurotrophin gene expression in human adipose-derived stem cells in vitro. J Adv Pharm Technol Res 2024; 15:214-219. [PMID: 39290551 PMCID: PMC11404434 DOI: 10.4103/japtr.japtr_390_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 01/10/2024] [Accepted: 02/05/2024] [Indexed: 09/19/2024] Open
Abstract
Rosemary leaf extract, a well-known medicinal plant, can induce neurotrophin gene expression and proliferation in stem cells. Human adipose-derived stem cells (hASCs) with high proliferation and differentiation capacity are easily accessible and can be extracted with the least damage. This study evaluated the effect of rosemary extract (RE) on neurotrophin gene expression at 48 h postinduction in hASCs. hASCs were isolated from healthy female donors, aged 28-35 years, who had undergone abdominal liposuction. Passage-4 stem cells were cultured and treated with different doses of RE (from 30 to 70 µg/ml) containing 40% carnosic acid for 48 h. Reverse transcription-polymerase chain reaction was used to check the expression of neurotrophin genes. The expression of NTF3, NTF4, and nerve growth factor genes in cells treated with 40-60 µg/ml and the expression of GDNF in cells treated with 50-70 µg/ml of RE for 48 h showed a significant increase compared to cells cultured in serum-containing medium. However, different doses of RE showed no effect on brain-derived neurotrophic factor gene expression in the treated cells. RE (50, 60 µg/ml) leads to an increase of neurotrophin gene expression in hASCs as compared to routine cell culture. Hence, this protocol can be used to prepare ideal cell sources for cell therapy.
Collapse
Affiliation(s)
- Dhiya Altememy
- Department of Pharmaceutics, College of Pharmacy, Al-Zahraa University for Women, Karbala, Iraq
| | - Maryam Haji Ghasem Kashani
- Department of Cellular and Molecular Biology, School of Biology, Institute of Biological Sciences, Damghan University, Damghan, Iran
| | - Amirahmadi Fateme
- Department of Cellular and Molecular Biology, School of Biology, Institute of Biological Sciences, Damghan University, Damghan, Iran
| | - Pegah Khosravian
- Medical Plants Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| |
Collapse
|
24
|
Saberian E, Jenča A, Zafari Y, Jenča A, Petrášová A, Zare-Zardini H, Jenčová J. Scaffold Application for Bone Regeneration with Stem Cells in Dentistry: Literature Review. Cells 2024; 13:1065. [PMID: 38920693 PMCID: PMC11202130 DOI: 10.3390/cells13121065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 06/14/2024] [Accepted: 06/18/2024] [Indexed: 06/27/2024] Open
Abstract
Bone tissue injuries within oral and dental contexts often present considerable challenges because traditional treatments may not be able to fully restore lost or damaged bone tissue. Novel approaches involving stem cells and targeted 3D scaffolds have been investigated in the search for workable solutions. The use of scaffolds in stem cell-assisted bone regeneration is a crucial component of tissue engineering techniques designed to overcome the drawbacks of traditional bone grafts. This study provides a detailed review of scaffold applications for bone regeneration with stem cells in dentistry. This review focuses on scaffolds and stem cells while covering a broad range of studies explaining bone regeneration in dentistry through the presentation of studies conducted in this field. The role of different stem cells in regenerative medicine is covered in great detail in the reviewed literature. These studies have addressed a wide range of subjects, including the effects of platelet concentrates during dental surgery or specific combinations, such as human dental pulp stem cells with scaffolds for animal model bone regeneration, to promote bone regeneration in animal models. Noting developments, research works consider methods to improve vascularization and explore the use of 3D-printed scaffolds, secretome applications, mesenchymal stem cells, and biomaterials for oral bone tissue regeneration. This thorough assessment outlines possible developments within these crucial regenerative dentistry cycles and provides insights and suggestions for additional study. Furthermore, alternative creative methods for regenerating bone tissue include biophysical stimuli, mechanical stimulation, magnetic field therapy, laser therapy, nutritional supplements and diet, gene therapy, and biomimetic materials. These innovative approaches offer promising avenues for future research and development in the field of bone tissue regeneration in dentistry.
Collapse
Affiliation(s)
- Elham Saberian
- Klinika of Stomatology and Maxillofacial Surgery Akadémia Košice Bacikova, Pavol Jozef Šafárik University, 040 01 Kosice, Slovakia
| | - Andrej Jenča
- Klinika of Stomatology and Maxillofacial Surgery Akadémia Košice Bacikova, Pavol Jozef Šafárik University, 040 01 Kosice, Slovakia
| | - Yaser Zafari
- Lyles School of Civil Engineering, Purdue University, West Lafayette, IN 47907, USA
| | - Andrej Jenča
- Klinika of Stomatology and Maxillofacial Surgery Akadémia Košice Bacikova, Pavol Jozef Šafárik University, 040 01 Kosice, Slovakia
| | - Adriána Petrášová
- Klinika of Stomatology and Maxillofacial Surgery Akadémia Košice Bacikova, Pavol Jozef Šafárik University, 040 01 Kosice, Slovakia
| | - Hadi Zare-Zardini
- Department of Biomedical Engineering, Meybod University, Meybod 89616-99557, Iran
| | - Janka Jenčová
- Klinika of Stomatology and Maxillofacial Surgery Akadémia Košice Bacikova, Pavol Jozef Šafárik University, 040 01 Kosice, Slovakia
| |
Collapse
|
25
|
Zhang S, Lee Y, Liu Y, Yu Y, Han I. Stem Cell and Regenerative Therapies for the Treatment of Osteoporotic Vertebral Compression Fractures. Int J Mol Sci 2024; 25:4979. [PMID: 38732198 PMCID: PMC11084822 DOI: 10.3390/ijms25094979] [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: 04/09/2024] [Revised: 04/28/2024] [Accepted: 04/30/2024] [Indexed: 05/13/2024] Open
Abstract
Osteoporotic vertebral compression fractures (OVCFs) significantly increase morbidity and mortality, presenting a formidable challenge in healthcare. Traditional interventions such as vertebroplasty and kyphoplasty, despite their widespread use, are limited in addressing the secondary effects of vertebral fractures in adjacent areas and do not facilitate bone regeneration. This review paper explores the emerging domain of regenerative therapies, spotlighting stem cell therapy's transformative potential in OVCF treatment. It thoroughly describes the therapeutic possibilities and mechanisms of action of mesenchymal stem cells against OVCFs, relying on recent clinical trials and preclinical studies for efficacy assessment. Our findings reveal that stem cell therapy, particularly in combination with scaffolding materials, holds substantial promise for bone regeneration, spinal stability improvement, and pain mitigation. This integration of stem cell-based methods with conventional treatments may herald a new era in OVCF management, potentially improving patient outcomes. This review advocates for accelerated research and collaborative efforts to translate laboratory breakthroughs into clinical practice, emphasizing the revolutionary impact of regenerative therapies on OVCF management. In summary, this paper positions stem cell therapy at the forefront of innovation for OVCF treatment, stressing the importance of ongoing research and cross-disciplinary collaboration to unlock its full clinical potential.
Collapse
Affiliation(s)
- Songzi Zhang
- Department of Neurosurgery, CHA Bundang Medical Center, CHA University, Seongnam-si 13496, Republic of Korea; (S.Z.); (Y.L.); (Y.Y.)
| | - Yunhwan Lee
- Department of Medicine, School of Medicine, CHA University, Seongnam-si 13496, Republic of Korea;
| | - Yanting Liu
- Department of Neurosurgery, CHA Bundang Medical Center, CHA University, Seongnam-si 13496, Republic of Korea; (S.Z.); (Y.L.); (Y.Y.)
| | - Yerin Yu
- Department of Neurosurgery, CHA Bundang Medical Center, CHA University, Seongnam-si 13496, Republic of Korea; (S.Z.); (Y.L.); (Y.Y.)
| | - Inbo Han
- Department of Neurosurgery, CHA Bundang Medical Center, CHA University, Seongnam-si 13496, Republic of Korea; (S.Z.); (Y.L.); (Y.Y.)
| |
Collapse
|
26
|
Wang S, Jia Z, Dai M, Feng X, Tang C, Liu L, Cao L. Advances in natural and synthetic macromolecules with stem cells and extracellular vesicles for orthopedic disease treatment. Int J Biol Macromol 2024; 268:131874. [PMID: 38692547 DOI: 10.1016/j.ijbiomac.2024.131874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Revised: 04/16/2024] [Accepted: 04/24/2024] [Indexed: 05/03/2024]
Abstract
Serious orthopedic disorders resulting from myriad diseases and impairments continue to pose a considerable challenge to contemporary clinical care. Owing to its limited regenerative capacity, achieving complete bone tissue regeneration and complete functional restoration has proven challenging with existing treatments. By virtue of cellular regenerative and paracrine pathways, stem cells are extensively utilized in the restoration and regeneration of bone tissue; however, low survival and retention after transplantation severely limit their therapeutic effect. Meanwhile, biomolecule materials provide a delivery platform that improves stem cell survival, increases retention, and enhances therapeutic efficacy. In this review, we present the basic concepts of stem cells and extracellular vesicles from different sources, emphasizing the importance of using appropriate expansion methods and modification strategies. We then review different types of biomolecule materials, focusing on their design strategies. Moreover, we summarize several forms of biomaterial preparation and application strategies as well as current research on biomacromolecule materials loaded with stem cells and extracellular vesicles. Finally, we present the challenges currently impeding their clinical application for the treatment of orthopedic diseases. The article aims to provide researchers with new insights for subsequent investigations.
Collapse
Affiliation(s)
- Supeng Wang
- The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou 325200, China; Jiujiang City Key Laboratory of Cell Therapy, The First Hospital of Jiujiang City, Jiujiang 332000, China; Ningxia Medical University, Ningxia 750004, China
| | - Zhiqiang Jia
- The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou 325200, China
| | - Minghai Dai
- The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou 325200, China
| | - Xujun Feng
- Jiujiang City Key Laboratory of Cell Therapy, The First Hospital of Jiujiang City, Jiujiang 332000, China
| | - Chengxuan Tang
- The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou 325200, China
| | - Liangle Liu
- The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou 325200, China.
| | - Lingling Cao
- Jiujiang City Key Laboratory of Cell Therapy, The First Hospital of Jiujiang City, Jiujiang 332000, China.
| |
Collapse
|
27
|
Yao J, Xin R, Zhao C, Yu C. MicroRNAs in osteoblast differentiation and fracture healing: From pathogenesis to therapeutic implication. Injury 2024; 55:111410. [PMID: 38359711 DOI: 10.1016/j.injury.2024.111410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 01/20/2024] [Accepted: 01/27/2024] [Indexed: 02/17/2024]
Abstract
The term "fracture" pertains to the occurrence of bones being either fully or partially disrupted as a result of external forces. Prolonged fracture healing can present a notable danger to the patient's general health and overall quality of life. The significance of osteoblasts in the process of new bone formation is widely recognized, and optimizing their function could be a desirable strategy. Therefore, the mending of bone fractures is intricately linked to the processes of osteogenic differentiation and mineralization. MicroRNAs (miRNAs) are RNA molecules that do not encode for proteins, but rather modulate the functioning of physiological processes by directly targeting proteins. The participation of microRNAs (miRNAs) in experimental investigations has been extensive, and their control functions have earned them the recognition as primary regulators of the human genome. Earlier studies have shown that modulating the expression of miRNAs, either by increasing or decreasing their levels, can initiate the differentiation of osteoblasts. This implies that miRNAs play a pivotal function in promoting osteogenesis, facilitating bone mineralization and formation, ultimately leading to an efficient healing of fractures. Hence, focusing on miRNAs can be considered a propitious therapeutic approach to accelerate the healing of fractures and forestall nonunion. In this manner, the information supplied by this investigation has the potential to aid in upcoming clinical utilization, including its possible use as biomarkers or as resources for devising innovative therapeutic tactics aimed at promoting fracture healing.
Collapse
Affiliation(s)
- Jilong Yao
- Department of surgery teaching and research section, Jiangxi Medical College, Shangrao, 334000, China
| | - Ruiwen Xin
- Department of surgery teaching and research section, Jiangxi Medical College, Shangrao, 334000, China
| | - Chao Zhao
- Department of Neurology, Shangrao municipal hospital, Shangrao, 334000, China
| | - Chunfu Yu
- Department of Neurology, Shangrao municipal hospital, Shangrao, 334000, China.
| |
Collapse
|
28
|
Rintanalert D, Ishida Y, Huang ACS, Hatano-Sato K, Li K, Chantarawaratit PO, Usumi-Fujita R, Hosomichi J, Ono T. SDF-1 involvement in orthodontic tooth movement after tooth extraction. Sci Rep 2024; 14:5048. [PMID: 38424199 PMCID: PMC10904391 DOI: 10.1038/s41598-024-55632-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 02/26/2024] [Indexed: 03/02/2024] Open
Abstract
The stromal cell-derived factor 1 (SDF-1)/chemokine receptor type 4 (CXCR4) axis plays a key role in alveolar bone metabolism during orthodontic tooth movement (OTM). Herein, the effects of the SDF-1/CXCR4 axis on the regional acceleratory phenomenon (RAP) in OTM velocity and on changes in the surrounding periodontium after adjacent tooth extraction in rats were investigated. Six-week-old male Wistar/ST rats underwent left maxillary first molar (M1) extraction and mesial OTM of the left maxillary second molar (M2) with a 10-g force closed-coil spring. Phosphate-buffered saline, immunoglobulin G (IgG) isotype control antibody, or anti-SDF-1 neutralizing monoclonal antibody were injected at the M1 and M2 interproximal areas (10 μg/0.1 mL) for the first three days. Analyses were performed after 1, 3, and 7 days (n = 7). The results demonstrated a significant increase in SDF-1 expression from day 1, which was effectively blocked via anti-SDF-1 neutralizing monoclonal antibody injection. On day 3, the M2 OTM distance and the number of positively stained osteoclasts significantly reduced alongside a reduction in inflammatory markers in the experimental group. Our results demonstrated that serial local injection of the anti-SDF-1 neutralizing monoclonal antibody reduces M2 OTM, osteoclast accumulation, and localized inflammatory responses in an OTM model with tooth extraction-induced RAP.
Collapse
Affiliation(s)
- Duangtawan Rintanalert
- Department of Orthodontic Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Yushima 1-5-45, Bunkyo-ku, Tokyo, 113-8510, Japan
- Department of Orthodontics, Faculty of Dentistry, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Yuji Ishida
- Department of Orthodontic Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Yushima 1-5-45, Bunkyo-ku, Tokyo, 113-8510, Japan.
| | - Albert Chun-Shuo Huang
- Department of Orthodontic Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Yushima 1-5-45, Bunkyo-ku, Tokyo, 113-8510, Japan
| | - Kasumi Hatano-Sato
- Department of Orthodontic Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Yushima 1-5-45, Bunkyo-ku, Tokyo, 113-8510, Japan
| | - Kai Li
- Department of Orthodontic Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Yushima 1-5-45, Bunkyo-ku, Tokyo, 113-8510, Japan
| | - Pintu-On Chantarawaratit
- Department of Orthodontics, Faculty of Dentistry, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Risa Usumi-Fujita
- Department of Orthodontic Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Yushima 1-5-45, Bunkyo-ku, Tokyo, 113-8510, Japan
| | - Jun Hosomichi
- Department of Orthodontic Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Yushima 1-5-45, Bunkyo-ku, Tokyo, 113-8510, Japan
| | - Takashi Ono
- Department of Orthodontic Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Yushima 1-5-45, Bunkyo-ku, Tokyo, 113-8510, Japan
| |
Collapse
|
29
|
Lu W, Shi Y, Qian M. Notoginsenoside R1 promotes osteogenic differentiation of human bone marrow mesenchymal stem cells via ERα/GSK-3β/β-catenin signalling pathway. Int J Exp Pathol 2024; 105:4-12. [PMID: 37899670 PMCID: PMC10797438 DOI: 10.1111/iep.12494] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 08/28/2023] [Accepted: 09/12/2023] [Indexed: 10/31/2023] Open
Abstract
Human bone marrow mesenchymal stem cells (hBMSCs) are attractive therapeutic agents for bone tissue regeneration owing to their osteogenic differentiation potential. Notoginsenoside R1 (NGR1) is a novel phytoestrogen with diverse pharmacological activities. Here, we probed whether NGR1 has an effect on the osteogenic differentiation of hBMSCs. EdU, CCK-8 and Transwell assays were used to measure proliferation and migration of hBMSCs after treatment with different doses of NGR1. hBMSCs were treated with osteogenic differentiation induction medium for osteogenesis. Alizarin red S (ARS) and alkaline phosphatase (ALP) staining were used to measure mineralized nodule formation and ALP activity in hBMSCs, respectively. ICI 182780, an antagonist of oestrogen receptor alpha (ERα) was used to inhibit ERα expression. The results showed that NGR1 enhanced hBMSC proliferation and migration. NGR1 increased ALP activity and mineralized nodule formation as well as promoting ALP, RUNX2 and OCN expression in hBMSCs. NGR1 enhanced ERα expression and promoted GSK-3β/β-catenin signal transduction in hBMSCs. ICI 182780 reversed NGR1-mediated activation of the GSK-3β/β-catenin signalling and promoted an effect on hBMSC behaviour. Thus NGR1 promotes proliferation, migration and osteogenic differentiation of hBMSCs via the ERα/GSK-3β/β-catenin signalling pathway.
Collapse
Affiliation(s)
- Wei Lu
- Department of TraumatologyChangshu No.2 People's HospitalJiangsuChina
| | - Yuanxin Shi
- Department of TraumatologyChangshu No.2 People's HospitalJiangsuChina
| | - Minglei Qian
- Department of TraumatologyChangshu No.2 People's HospitalJiangsuChina
| |
Collapse
|
30
|
Wilson BJ, Owston HE, Iqbal N, Giannoudis PV, McGonagle D, Pandit H, Philipose Pampadykandathil L, Jones E, Ganguly P. In Vitro Osteogenesis Study of Shell Nacre Cement with Older and Young Donor Bone Marrow Mesenchymal Stem/Stromal Cells. Bioengineering (Basel) 2024; 11:143. [PMID: 38391629 PMCID: PMC10886325 DOI: 10.3390/bioengineering11020143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Revised: 01/22/2024] [Accepted: 01/26/2024] [Indexed: 02/24/2024] Open
Abstract
Bone void-filling cements are one of the preferred materials for managing irregular bone voids, particularly in the geriatric population who undergo many orthopedic surgeries. However, bone marrow mesenchymal stem/stromal cells (BM-MSCs) of older-age donors often exhibit reduced osteogenic capacity. Hence, it is crucial to evaluate candidate bone substitute materials with BM-MSCs from the geriatric population to determine the true osteogenic potential, thus simulating the clinical situation. With this concept, we investigated the osteogenic potential of shell nacre cement (SNC), a bone void-filling cement based on shell nacre powder and ladder-structured siloxane methacrylate, using older donor BM-MSCs (age > 55 years) and young donor BM-MSCs (age < 30 years). Direct and indirect cytotoxicity studies conducted with human BM-MSCs confirmed the non-cytotoxic nature of SNC. The standard colony-forming unit-fibroblast (CFU-F) assay and population doubling (PD) time assays revealed a significant reduction in the proliferation potential (p < 0.0001, p < 0.05) in older donor BM-MSCs compared to young donor BM-MSCs. Correspondingly, older donor BM-MSCs contained higher proportions of senescent, β-galactosidase (SA-β gal)-positive cells (nearly 2-fold, p < 0.001). In contrast, the proliferation capacity of older donor BM-MSCs, measured as the area density of CellTrackerTM green positive cells, was similar to that of young donor BM-MSCs following a 7-day culture on SNC. Furthermore, after 14 days of osteoinduction on SNC, scanning electron microscopy with energy-dispersive spectroscopy (SEM-EDS) showed that the amount of calcium and phosphorus deposited by young and older donor BM-MSCs on SNC was comparable. A similar trend was observed in the expression of the osteogenesis-related genes BMP2, RUNX2, ALP, COL1A1, OMD and SPARC. Overall, the results of this study indicated that SNC would be a promising candidate for managing bone voids in all age groups.
Collapse
Affiliation(s)
- Bridget Jeyatha Wilson
- Division of Dental Products, Department of Biomaterial Science and Technology, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Thiruvananthapuram 695 012, India
| | - Heather Elizabeth Owston
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds LS9 7JT, UK (D.M.); (H.P.)
| | - Neelam Iqbal
- School of Chemical and Process Engineering, University of Leeds, Leeds LS2 9JT, UK
| | - Peter V. Giannoudis
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds LS9 7JT, UK (D.M.); (H.P.)
- Leeds Orthopaedic & Trauma Sciences, Leeds General Infirmary, University of Leeds, Leeds LS2 9JT, UK
- Leeds Musculoskeletal Biomedical Research Centre, Chapel Allerton Hospital, Leeds LS7 4SA, UK
| | - Dennis McGonagle
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds LS9 7JT, UK (D.M.); (H.P.)
- Leeds Musculoskeletal Biomedical Research Centre, Chapel Allerton Hospital, Leeds LS7 4SA, UK
| | - Hemant Pandit
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds LS9 7JT, UK (D.M.); (H.P.)
- Leeds Musculoskeletal Biomedical Research Centre, Chapel Allerton Hospital, Leeds LS7 4SA, UK
| | - Lizymol Philipose Pampadykandathil
- Division of Dental Products, Department of Biomaterial Science and Technology, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Thiruvananthapuram 695 012, India
| | - Elena Jones
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds LS9 7JT, UK (D.M.); (H.P.)
| | - Payal Ganguly
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds LS9 7JT, UK (D.M.); (H.P.)
| |
Collapse
|
31
|
Huang F, Wei G, Wang H, Zhang Y, Lan W, Xie Y, Wu G. Fibroblasts inhibit osteogenesis by regulating nuclear-cytoplasmic shuttling of YAP in mesenchymal stem cells and secreting DKK1. Biol Res 2024; 57:4. [PMID: 38245803 PMCID: PMC10799393 DOI: 10.1186/s40659-023-00481-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Accepted: 12/04/2023] [Indexed: 01/22/2024] Open
Abstract
BACKGROUND Fibrous scars frequently form at the sites of bone nonunion when attempts to repair bone fractures have failed. However, the detailed mechanism by which fibroblasts, which are the main components of fibrous scars, impede osteogenesis remains largely unknown. RESULTS In this study, we found that fibroblasts compete with osteogenesis in both human bone nonunion tissues and BMP2-induced ectopic osteogenesis in a mouse model. Fibroblasts could inhibit the osteoblastic differentiation of mesenchymal stem cells (MSCs) via direct and indirect cell competition. During this process, fibroblasts modulated the nuclear-cytoplasmic shuttling of YAP in MSCs. Knocking down YAP could inhibit osteoblast differentiation of MSCs, while overexpression of nuclear-localized YAP-5SA could reverse the inhibition of osteoblast differentiation of MSCs caused by fibroblasts. Furthermore, fibroblasts secreted DKK1, which further inhibited the formation of calcium nodules during the late stage of osteogenesis but did not affect the early stage of osteogenesis. Thus, fibroblasts could inhibit osteogenesis by regulating YAP localization in MSCs and secreting DKK1. CONCLUSIONS Our research revealed that fibroblasts could modulate the nuclear-cytoplasmic shuttling of YAP in MSCs, thereby inhibiting their osteoblast differentiation. Fibroblasts could also secrete DKK1, which inhibited calcium nodule formation at the late stage of osteogenesis.
Collapse
Affiliation(s)
- Fei Huang
- Central Laboratory, First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, Fujian, China
| | - Guozhen Wei
- Department of Orthopaedics, The First Affiliated Hospital, Fujian Medical University, No. 20, Chazhong Road, Taijiang District, Fuzhou, 350005, Fujian, China
- Department of Orthopaedics, Binhai Campus of the First Affiliated Hospital, National Regional Medical Center, Fujian Medical University, Fuzhou, 350212, Fujian, China
| | - Hai Wang
- Department of Orthopaedics, The First Affiliated Hospital, Fujian Medical University, No. 20, Chazhong Road, Taijiang District, Fuzhou, 350005, Fujian, China
- Department of Orthopaedics, Binhai Campus of the First Affiliated Hospital, National Regional Medical Center, Fujian Medical University, Fuzhou, 350212, Fujian, China
| | - Ying Zhang
- Central Laboratory, First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, Fujian, China
| | - Wenbin Lan
- Department of Orthopaedics, The First Affiliated Hospital, Fujian Medical University, No. 20, Chazhong Road, Taijiang District, Fuzhou, 350005, Fujian, China
- Department of Orthopaedics, Binhai Campus of the First Affiliated Hospital, National Regional Medical Center, Fujian Medical University, Fuzhou, 350212, Fujian, China
| | - Yun Xie
- Department of Orthopaedics, The First Affiliated Hospital, Fujian Medical University, No. 20, Chazhong Road, Taijiang District, Fuzhou, 350005, Fujian, China.
- Department of Orthopaedics, Binhai Campus of the First Affiliated Hospital, National Regional Medical Center, Fujian Medical University, Fuzhou, 350212, Fujian, China.
| | - Gui Wu
- Department of Orthopaedics, The First Affiliated Hospital, Fujian Medical University, No. 20, Chazhong Road, Taijiang District, Fuzhou, 350005, Fujian, China.
- Department of Orthopaedics, Binhai Campus of the First Affiliated Hospital, National Regional Medical Center, Fujian Medical University, Fuzhou, 350212, Fujian, China.
| |
Collapse
|
32
|
Keum BR, Kim HJ, Kim GH, Chang DG. Osteobiologies for Spinal Fusion from Biological Mechanisms to Clinical Applications: A Narrative Review. Int J Mol Sci 2023; 24:17365. [PMID: 38139194 PMCID: PMC10743675 DOI: 10.3390/ijms242417365] [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: 11/17/2023] [Revised: 12/01/2023] [Accepted: 12/03/2023] [Indexed: 12/24/2023] Open
Abstract
Degenerative lumbar spinal disease (DLSD), including spondylolisthesis and spinal stenosis, is increasing due to the aging population. Along with the disease severity, lumbar interbody fusion (LIF) is a mainstay of surgical treatment through decompression, the restoration of intervertebral heights, and the stabilization of motion segments. Currently, pseudoarthrosis after LIF is an important and unsolved issue, which is closely related to osteobiologies. Of the many signaling pathways, the bone morphogenetic protein (BMP) signaling pathway contributes to osteoblast differentiation, which is generally regulated by SMAD proteins as common in the TGF-β superfamily. BMP-2 and -4 are also inter-connected with Wnt/β-catenin, Notch, and FGF signaling pathways. With the potent potential for osteoinduction in BMP-2 and -4, the combination of allogenous bone and recombinant human BMPs (rhBMPs) is currently an ideal fusion material, which has equalized or improved fusion rates compared to traditional materials. However, safety issues in the dosage of BMP remain, so overcoming current limitations will provide significant advancement in spine surgery. In the future, translational research and the application of clinical study will be important to overcome the current limitations of spinal surgery.
Collapse
Affiliation(s)
- Byeong-Rak Keum
- Research Center for Bioconvergence Analysis, Korea Basic Science Institute, Cheongju 28119, Republic of Korea;
| | - Hong Jin Kim
- Department of Orthopedic Surgery, Inje University Sanggye Paik Hospital, College of Medicine, Inje University, Seoul 01757, Republic of Korea;
| | - Gun-Hwa Kim
- Research Center for Bioconvergence Analysis, Korea Basic Science Institute, Cheongju 28119, Republic of Korea;
| | - Dong-Gune Chang
- Department of Orthopedic Surgery, Inje University Sanggye Paik Hospital, College of Medicine, Inje University, Seoul 01757, Republic of Korea;
| |
Collapse
|
33
|
Wang S, Liu Y, Lou C, Cai C, Ren W, Liu J, Gong M, Shang P, Zhang H. Moderate static magnetic field promotes fracture healing and regulates iron metabolism in mice. Biomed Eng Online 2023; 22:107. [PMID: 37968671 PMCID: PMC10647027 DOI: 10.1186/s12938-023-01170-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 10/31/2023] [Indexed: 11/17/2023] Open
Abstract
BACKGROUND Fractures are the most common orthopedic diseases. It is known that static magnetic fields (SMFs) can contribute to the maintenance of bone health. However, the effect and mechanism of SMFs on fracture is still unclear. This study is aim to investigate the effect of moderate static magnetic fields (MMFs) on bone structure and metabolism during fracture healing. METHODS Eight-week-old male C57BL/6J mice were subjected to a unilateral open transverse tibial fracture, and following treatment under geomagnetic field (GMF) or MMF. The micro-computed tomography (Micro-CT) and three-point bending were employed to evaluate the microarchitecture and mechanical properties. Endochondral ossification and bone remodeling were evaluated by bone histomorphometric and serum biochemical assay. In addition, the atomic absorption spectroscopy and ELISA were utilized to examine the influence of MMF exposure on iron metabolism in mice. RESULTS MMF exposure increased bone mineral density (BMD), bone volume per tissue volume (BV/TV), mechanical properties, and proportion of mineralized bone matrix of the callus during fracture healing. MMF exposure reduced the proportion of cartilage in the callus area during fracture healing. Meanwhile, MMF exposure increased the number of osteoblasts in callus on the 14th day, and reduced the number of osteoclasts on the 28th day of fracture healing. Furthermore, MMF exposure increased PINP and OCN levels, and reduced the TRAP-5b and β-CTX levels in serum. It was also observed that MMF exposure reduced the iron content in the liver and callus, as well as serum ferritin levels while elevating the serum hepcidin concentration. CONCLUSIONS MMF exposure could accelerate fracture healing via promote the endochondral ossification and bone formation while regulating systemic iron metabolism during fracture healing. This study suggests that MMF may have the potential to become a form of physical therapy for fractures.
Collapse
Affiliation(s)
- Shenghang Wang
- Department of Spine Surgery, People's Hospital of Longhua, Affiliated Hospital of Southern Medical University, No.38 Jinglong Construction Road, Shenzhen, China
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Yuetong Liu
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
- Research & Development Institute of Northwestern Polytechnical University in Shenzhen, No. 45, Gaoxin South 9th Road, Nanshan District, Shenzhen, 518057, China
| | - Chenge Lou
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
- Research & Development Institute of Northwestern Polytechnical University in Shenzhen, No. 45, Gaoxin South 9th Road, Nanshan District, Shenzhen, 518057, China
| | - Chao Cai
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
- Research & Development Institute of Northwestern Polytechnical University in Shenzhen, No. 45, Gaoxin South 9th Road, Nanshan District, Shenzhen, 518057, China
| | - Weihao Ren
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
- Research & Development Institute of Northwestern Polytechnical University in Shenzhen, No. 45, Gaoxin South 9th Road, Nanshan District, Shenzhen, 518057, China
| | - Junyu Liu
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
- Research & Development Institute of Northwestern Polytechnical University in Shenzhen, No. 45, Gaoxin South 9th Road, Nanshan District, Shenzhen, 518057, China
| | - Ming Gong
- Department of Spine Surgery, People's Hospital of Longhua, Affiliated Hospital of Southern Medical University, No.38 Jinglong Construction Road, Shenzhen, China
| | - Peng Shang
- Research & Development Institute of Northwestern Polytechnical University in Shenzhen, No. 45, Gaoxin South 9th Road, Nanshan District, Shenzhen, 518057, China.
| | - Hao Zhang
- Department of Spine Surgery, People's Hospital of Longhua, Affiliated Hospital of Southern Medical University, No.38 Jinglong Construction Road, Shenzhen, China.
| |
Collapse
|
34
|
Kyriakaki I, Karanikola T, Lillis T, Kontonasaki E, Dabarakis N. Effect of direct oral anticoagulant dabigatran on early bone healing: An experimental study in rats. JOURNAL OF ADVANCED PERIODONTOLOGY & IMPLANT DENTISTRY 2023; 15:86-92. [PMID: 38357331 PMCID: PMC10862050 DOI: 10.34172/japid.2023.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 09/30/2023] [Indexed: 02/16/2024]
Abstract
Background Dabigatran belongs to the new generation of direct oral anticoagulants (DOACs). Its advantages are oral administration and no need for international normalized ratio (INR) monitoring. Although its use has increased, its potential side effects on bone healing and remodeling have not been fully investigated. The present study aimed to evaluate the possible effects of dabigatran on early bone healing. Methods Sixteen male Wistar rats were divided into two groups; in group A, 20-mg/kg dabigatran dose was administered orally daily for 15 days, while group B served as a control. Two circular bone defects (d=6 mm) were created on either side of the parietal bones. Two weeks after surgery and euthanasia of the animals, tissue samples (parietal bones that contained the defects) were harvested for histological and histomorphometric analysis. Statistical analysis was performed with a significance level of α=0.5. Results No statistically significant differences were found between the two groups regarding the regenerated bone (21.9% vs. 16.3%, P=0.172) or the percentage of bone bridging (63.3% vs. 53.5%, P=0.401). Conclusion Dabigatran did not affect bone regeneration, suggesting that it might be a safer drug compared to older anticoagulants known to lead to bone healing delay.
Collapse
Affiliation(s)
- Ioanna Kyriakaki
- Department of Dentoalveolar Surgery, Surgical Implantology and Roentgenology, Aristotle University, Thessaloniki, Greece
| | - Theodora Karanikola
- Private Practice, Clinical Instructor, Department of Oral Surgery, Implantology and Dental Radiology, School of Dentistry, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Theodoros Lillis
- Department of Dentoalveolar Surgery, Surgical Implantology and Roentgenology, Aristotle University, Thessaloniki, Greece
| | - Eleana Kontonasaki
- Department of Prosthodontics, School of Dentistry, Aristotle University of Thessaloniki, Greece
| | - Nikolaos Dabarakis
- Department of Dentoalveolar Surgery, Surgical Implantology and Roentgenology, Aristotle University, Thessaloniki, Greece
| |
Collapse
|
35
|
Tang H, Li J, Li JK, He SH, Xiang G, Rong R, Liang ZT, Zhang HQ. BMP6 participates in the pathogenesis of adolescent idiopathic scoliosis by regulating osteopenia. J Cell Physiol 2023; 238:2586-2599. [PMID: 37795636 DOI: 10.1002/jcp.31111] [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: 05/22/2023] [Revised: 08/08/2023] [Accepted: 08/17/2023] [Indexed: 10/06/2023]
Abstract
Adolescent idiopathic scoliosis (AIS) is a complex disease characterized by three-dimensional structural deformities of the spine. Its pathogenesis is associated with osteopenia. Bone-marrow-derived mesenchymal stem cells (BMSCs) play an important role in bone metabolism. We detected 1919 differentially expressed mRNAs and 744 differentially expressed lncRNAs in BMSCs from seven patients with AIS and five patients without AIS via high-throughput sequencing. Multiple analyses identified bone morphogenetic protein-6 (BMP6) as a hub gene that regulates the abnormal osteogenic differentiation of BMSCs in AIS. BMP6 expression was found to be decreased in AIS and its knockdown in human BMSCs significantly altered the degree of osteogenic differentiation. Additionally, CAP1-217 has been shown to be a potential upstream regulatory molecule of BMP6. We showed that CAP1-217 knockdown downregulated the expression of BMP6 and the osteogenic differentiation of BMSCs. Simultaneously, knockout of BMP6 in zebrafish embryos significantly increased the deformity rate. The findings of this study suggest that BMP6 is a key gene that regulates the abnormal osteogenic differentiation of BMSCs in AIS via the CAP1-217/BMP6/RUNX2 axis.
Collapse
Affiliation(s)
- Hao Tang
- Department of Spine Surgery and Orthopaedics, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
| | - Jiong Li
- Department of Spine Surgery and Orthopaedics, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
| | - Jia-Ke Li
- Department of General Surgery, The Third Xiangya Hospital of Central South University, Changsha, People's Republic of China
| | - Si-Han He
- Department of Spine Surgery and Orthopaedics, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
| | - Gang Xiang
- Department of Spine Surgery and Orthopaedics, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
| | - Rong Rong
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
- Hunan Key Laboratory of Ophthalmology, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
| | - Zhuo-Tao Liang
- Department of Spine Surgery and Orthopaedics, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
| | - Hong-Qi Zhang
- Department of Spine Surgery and Orthopaedics, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
| |
Collapse
|
36
|
Histological evaluation of the effects of bone morphogenetic protein 9 and angiopoietin 1 on bone healing. J Taibah Univ Med Sci 2023; 18:954-963. [PMID: 36875339 PMCID: PMC9982621 DOI: 10.1016/j.jtumed.2023.02.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Revised: 01/09/2023] [Accepted: 02/08/2023] [Indexed: 02/24/2023] Open
Abstract
Objectives Bone healing remains a critical clinical orthopedic problem. Bone, which is a greatly vascularized tissue, depends on the tight temporal and spatial link between blood vessels and bone cells. Thus, angiogenesis is crucial for skeletal growth and bone fracture healing. The purpose of this study was to evaluate the efficacy of the local application of osteogenic and angiogenic factors such as bone morphogenetic protein 9 (BMP9) and angiopoietin 1 (Ang1), respectively, and their combination as an osteoinducer in the process of bone healing. Methods Forty-eight male albino rats, weighing 300-400 g and aged 6-8 months, were utilized in this study. The animals underwent surgery on the medial side of the tibia bone. In the control group, an absorbable hemostatic sponge was locally applied to the bone defect, while experimental groups were separated into three groups. In Group I, 1 mg BMP9 was locally applied, Group II was treated with 1 mg Ang1, and Group III was treated with local application of a combination (0.5 mg BMP9 and 0.5 mg Ang1). All experimental groups were fixed with an absorbable hemostatic sponge. The rats were sacrificed on days 14 and 28 after surgery. Results Local application of BMP9 alone, Ang1 alone, and their combination to a tibia defect caused osteoid tissue formation and significantly increased the number of bone cells. A gradual decrease in the number of trabecular bone, an increase in trabecular area, and no significant difference in the bone marrow area were noted. Conclusion The combination of BMP9 and Ang1 has therapeutic potential in promoting the healing process of bone defects. Osteogenesis and angiogenesis are regulated by BMP9 and Ang1. These factors act together to accelerate bone regeneration more efficiently than either factor alone.
Collapse
|
37
|
Liang H, Li Q, Wang N, Wang C, Shi S, Yang H, Cao Y, Shi R, Jin L, Zhang C. KDM4D enhances osteo/dentinogenic differentiation and migration of SCAPs via binding to RPS5. Oral Dis 2023; 29:2827-2836. [PMID: 36579641 DOI: 10.1111/odi.14479] [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: 06/19/2022] [Revised: 10/21/2022] [Accepted: 12/19/2022] [Indexed: 12/30/2022]
Abstract
OBJECTIVES Stem cells of the apical papilla (SCAPs) provide promising candidates for dental pulp regeneration. Despite great advances in the transcriptional controls of the SCAPs fate, little is known about the regulation of SCAP differentiation. MATERIALS AND METHODS Short hairpin RNAs and full-length RNA were used to deplete or overexpress lysine demethylase 4D (KDM4D) gene expression. Western blotting, real-time RT-PCR, alizarin red staining, and scratch migration assays were used to study the role of KDM4D and the ribosomal protein encoded by RPS5 in SCAPs. RNA microarray, chromatin Immunoprecipitation (ChIP), and co-immunoprecipitation (Co-IP) assays were performed to explore the underlying molecular mechanisms. RESULTS KDM4D enhanced the osteo/dentinogenic differentiation, migration, and chemotaxis of SCAPs. The microarray results revealed that 88 mRNAs were differentially expressed in KDM4D-overexpressed SCAPs. ChIP results showed knock-down of KDM4D increased the level of H3K9me2 and H3K9me3 in CNR1 promoter region. There were 37 possible binding partners of KDM4D. KDM4D was found to combine with RPS5, which also promoted the osteo/dentinogenic differentiation, migration, and chemotaxis of SCAPs. CONCLUSIONS KDM4D promoted the osteo/dentinogenic differentiation and migration potential of SCAPs in combination with RPS5, which provides a therapeutic clue for improving SCAPs-based dental tissue regeneration.
Collapse
Affiliation(s)
- Hanbing Liang
- Laboratory of Molecular Signaling and Stem Cells Therapy, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, China
- Department of Endodontics, Capital Medical University School of Stomatology, Beijing, China
| | - Qian Li
- Laboratory of Molecular Signaling and Stem Cells Therapy, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, China
- Department of Endodontics, Capital Medical University School of Stomatology, Beijing, China
| | - Ning Wang
- Laboratory of Molecular Signaling and Stem Cells Therapy, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, China
| | - Chunxiong Wang
- Laboratory of Molecular Signaling and Stem Cells Therapy, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, China
- Department of Endodontics, Capital Medical University School of Stomatology, Beijing, China
| | - Shaojing Shi
- Laboratory of Molecular Signaling and Stem Cells Therapy, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, China
- Department of Endodontics, Capital Medical University School of Stomatology, Beijing, China
| | - Haoqing Yang
- Laboratory of Molecular Signaling and Stem Cells Therapy, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, China
| | - Yangyang Cao
- Laboratory of Molecular Signaling and Stem Cells Therapy, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, China
| | - Ruitang Shi
- Laboratory of Molecular Signaling and Stem Cells Therapy, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, China
- Department of Endodontics, Capital Medical University School of Stomatology, Beijing, China
| | - Luyuan Jin
- Department of General Dentistry and Integrated Emergency Dental Care, Capital Medical University School of Stomatology, Beijing, China
| | - Chen Zhang
- Department of Endodontics, Capital Medical University School of Stomatology, Beijing, China
| |
Collapse
|
38
|
Liu Q, Wu Y, Li S, Yoon S, Zhang J, Wang X, Hu L, Su C, Zhang C, Wu Y. Ursolic acid alleviates steroid-induced avascular necrosis of the femoral head in mouse by inhibiting apoptosis and rescuing osteogenic differentiation. Toxicol Appl Pharmacol 2023; 475:116649. [PMID: 37536651 DOI: 10.1016/j.taap.2023.116649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Revised: 07/19/2023] [Accepted: 07/31/2023] [Indexed: 08/05/2023]
Abstract
Steroid-induced avascular necrosis of femoral head (SANFH) is a common disorder worldwide with high disability. Overdose of glucocorticoid (GC) is the most common non-traumatic cause of SANFH. Up until now, there are limited therapeutic strategies for curing SANFH, and the mechanisms underlying SANFH progression remain unclear. Nevertheless, Osteogenic dysfunction is considered to be one of the crucial pathobiological mechanisms in the development of SANFH, which involves mouse bone marrow mesenchymal stem cells (BMSCs) apoptosis and osteogenic differentiation disorder. Ursolic acid (UA), an important component of the Chinese medicine formula Yougui Yin, has a wide range of pharmacological properties such as anti-tumor, anti-inflammatory and bone remodeling. Due to the positive effect of Yougui Yin on bone remodeling, the purpose of this study was to investigate the effects of UA on dexamethasone (DEX)-induced SANFH in vitro and vivo. In vitro, we demonstrated that UA can promote mouse BMSCs proliferation and resist DEX-induced apoptosis by CCK8, Western blotting, TUNEL and so on. In addition, vitro experiments such as ALP and Alizarin red staining assay showed that UA had a beneficial effect on the osteogenic differentiation of mouse BMSCs. In vivo, the results of H&E staining, immunohistochemistry staining, Elisa and micro-CT analysis showed that UA had a bone repair-promoting effect in SANFH model. Moreover, the results of Western blot and TUNEL experiments showed that UA could delay the disease progression of SANFH in mice by inhibiting apoptosis. Overall, our study suggests that UA is a potential compound for the treatment of SANFH.
Collapse
Affiliation(s)
- Qian Liu
- Department of the Orthopedics of Traditional Chinese Medicine, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China; College of Pharmacy, Chonnam National University, Gwangju, Republic of Korea; School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Yuyang Wu
- School of the 1st Clinical Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Sisi Li
- Department of Otolaryngology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Somy Yoon
- College of Pharmacy, Chonnam National University, Gwangju, Republic of Korea
| | - Jiaxin Zhang
- Department of the Orthopedics of Traditional Chinese Medicine, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xiaoyi Wang
- Department of the Orthopedics of Traditional Chinese Medicine, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Luoshuang Hu
- Department of the Orthopedics of Traditional Chinese Medicine, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Chenying Su
- Department of the Orthopedics of Traditional Chinese Medicine, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Chunwu Zhang
- Department of the Orthopedics of Traditional Chinese Medicine, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.
| | - Yungang Wu
- Department of the Orthopedics of Traditional Chinese Medicine, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.
| |
Collapse
|
39
|
Lau CS, Chua J, Prasadh S, Lim J, Saigo L, Goh BT. Alveolar Ridge Augmentation with a Novel Combination of 3D-Printed Scaffolds and Adipose-Derived Mesenchymal Stem Cells-A Pilot Study in Pigs. Biomedicines 2023; 11:2274. [PMID: 37626770 PMCID: PMC10452669 DOI: 10.3390/biomedicines11082274] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 08/01/2023] [Accepted: 08/10/2023] [Indexed: 08/27/2023] Open
Abstract
Alveolar ridge augmentation is an important dental procedure to increase the volume of bone tissue in the alveolar ridge before the installation of a dental implant. To meet the high demand for bone grafts for alveolar ridge augmentation and to overcome the limitations of autogenous bone, allografts, and xenografts, researchers are developing bone grafts from synthetic materials using novel fabrication techniques such as 3D printing. To improve the clinical performance of synthetic bone grafts, stem cells with osteogenic differentiation capability can be loaded into the grafts. In this pilot study, we propose a novel bone graft which combines a 3D-printed polycaprolactone-tricalcium phosphate (PCL-TCP) scaffold with adipose-derived mesenchymal stem cells (AD-MSCs) that can be harvested, processed and implanted within the alveolar ridge augmentation surgery. We evaluated the novel bone graft in a porcine lateral alveolar defect model. Radiographic analysis revealed that the addition of AD-MSCs to the PCL-TCP scaffold improved the bone volume in the defect from 18.6% to 28.7% after 3 months of healing. Histological analysis showed the presence of AD-MSCs in the PCL-TCP scaffold led to better formation of new bone and less likelihood of fibrous encapsulation of the scaffold. Our pilot study demonstrated that the loading of AD-MSCs improved the bone regeneration capability of PCL-TCP scaffolds, and our novel bone graft is suitable for alveolar ridge augmentation.
Collapse
Affiliation(s)
- Chau Sang Lau
- National Dental Research Institute Singapore, National Dental Centre Singapore, Singapore 168938, Singapore; (C.S.L.); (L.S.)
- Oral Health Academic Clinical Programme, Duke-NUS Medical School, Singapore 169857, Singapore
| | - Jasper Chua
- Duke-NUS Medical School, Singapore 169857, Singapore;
| | - Somasundaram Prasadh
- Center for Clean Energy Engineering, University of Connecticut, Storrs, CT 06269, USA;
| | - Jing Lim
- Osteopore International Pte Ltd., Singapore 618305, Singapore;
| | - Leonardo Saigo
- National Dental Research Institute Singapore, National Dental Centre Singapore, Singapore 168938, Singapore; (C.S.L.); (L.S.)
| | - Bee Tin Goh
- National Dental Research Institute Singapore, National Dental Centre Singapore, Singapore 168938, Singapore; (C.S.L.); (L.S.)
- Oral Health Academic Clinical Programme, Duke-NUS Medical School, Singapore 169857, Singapore
| |
Collapse
|
40
|
Jin Y, Li S, Yu Q, Chen T, Liu D. Application of stem cells in regeneration medicine. MedComm (Beijing) 2023; 4:e291. [PMID: 37337579 PMCID: PMC10276889 DOI: 10.1002/mco2.291] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 04/25/2023] [Accepted: 05/08/2023] [Indexed: 06/21/2023] Open
Abstract
Regeneration is a complex process affected by many elements independent or combined, including inflammation, proliferation, and tissue remodeling. Stem cells is a class of primitive cells with the potentiality of differentiation, regenerate with self-replication, multidirectional differentiation, and immunomodulatory functions. Stem cells and their cytokines not only inextricably linked to the regeneration of ectodermal and skin tissues, but also can be used for the treatment of a variety of chronic wounds. Stem cells can produce exosomes in a paracrine manner. Stem cell exosomes play an important role in tissue regeneration, repair, and accelerated wound healing, the biological properties of which are similar with stem cells, while stem cell exosomes are safer and more effective. Skin and bone tissues are critical organs in the body, which are essential for sustaining life activities. The weak repairing ability leads a pronounced impact on the quality of life of patients, which could be alleviated by stem cell exosomes treatment. However, there are obstacles that stem cells and stem cells exosomes trough skin for improved bioavailability. This paper summarizes the applications and mechanisms of stem cells and stem cells exosomes for skin and bone healing. We also propose new ways of utilizing stem cells and their exosomes through different nanoformulations, liposomes and nanoliposomes, polymer micelles, microspheres, hydrogels, and scaffold microneedles, to improve their use in tissue healing and regeneration.
Collapse
Affiliation(s)
- Ye Jin
- School of PharmacyChangchun University of Chinese MedicineChangchunJilinChina
| | - Shuangyang Li
- School of PharmacyChangchun University of Chinese MedicineChangchunJilinChina
| | - Qixuan Yu
- School of PharmacyChangchun University of Chinese MedicineChangchunJilinChina
| | - Tianli Chen
- School of PharmacyChangchun University of Chinese MedicineChangchunJilinChina
| | - Da Liu
- School of PharmacyChangchun University of Chinese MedicineChangchunJilinChina
| |
Collapse
|
41
|
Hu Z, Jiang Z, Meng S, Liu R, Yang K. Research Progress on the Osteogenesis-Related Regulatory Mechanisms of Human Umbilical Cord Mesenchymal Stem Cells. Stem Cell Rev Rep 2023; 19:1252-1267. [PMID: 36917312 DOI: 10.1007/s12015-023-10521-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/19/2023] [Indexed: 03/16/2023]
Abstract
In recent years, research on human umbilical cord mesenchymal stem cells (hUCMSCs) derived from human umbilical cord tissue has accelerated and entered clinical application research. Compared with mesenchymal stem cells (MSCs) from other sources, hUCMSCs can be extracted from different parts of umbilical cord or from the whole umbilical cord. It has the characteristics of less ethical controversy, high differentiation potential, strong proliferation ability, efficient expansion in vitro, avoiding immune rejection and immune privilege, and avoids the limitations of lack of embryonic stem cells, heterogeneity, ethical and moral constraints. hUCMSCs avoid the need for embryonic stem cell sources, heterogeneity, and ethical and moral constraints. Bone defects are very common in clinical practice, but completely effective bone tissue regeneration treatment is challenging. Currently, autologous bone transplantation and allogeneic bone transplantation are main treatment approaches in clinical work, but each has different shortcomings, such as limited sources, invasiveness, immune rejection and insufficient osteogenic ability. Therefore, to solve the bottleneck of bone tissue regeneration and repair, a great amount of research has been carried out to explore the clinical advantages of hUCMSCs as seed cells to promote osteogenesis.However, the regulation of osteogenic differentiation of hUCMSCs is an extremely complex process. Although a large number of studies have demonstrated that the role of hUCMSCs in enhancing local bone regeneration and repair through osteogenic differentiation and transplantation into the body involves multiple signaling pathways, there is no relevant article that summarize the findings. This article discusses the osteogenesis-related regulatory mechanisms of hUCMSCs, summarizes the currently known related mechanisms, and speculates on the possible signals.
Collapse
Affiliation(s)
- Zhengqi Hu
- Department of Periodontology, Hospital of Stomatology, Zunyi Medical University, Zunyi, 563000, Guizhou, China
| | - Zhiliang Jiang
- Department of Periodontology, Hospital of Stomatology, Zunyi Medical University, Zunyi, 563000, Guizhou, China
| | - Shengzi Meng
- Department of Periodontology, Hospital of Stomatology, Zunyi Medical University, Zunyi, 563000, Guizhou, China
| | - Rong Liu
- Department of Periodontology, Hospital of Stomatology, Zunyi Medical University, Zunyi, 563000, Guizhou, China
| | - Kun Yang
- Department of Periodontology, Hospital of Stomatology, Zunyi Medical University, Zunyi, 563000, Guizhou, China.
| |
Collapse
|
42
|
Pennasilico L, Di Bella C, Sassaroli S, Salvaggio A, Roggiolani F, Piccionello AP. Effects of Autologous Microfragmented Adipose Tissue on Healing of Tibial Plateau Levelling Osteotomies in Dogs: A Prospective Clinical Trial. Animals (Basel) 2023; 13:2084. [PMID: 37443881 DOI: 10.3390/ani13132084] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 06/20/2023] [Accepted: 06/21/2023] [Indexed: 07/15/2023] Open
Abstract
The aim of this study was to evaluate the effects of autologous microfragmented adipose tissue (MFAT) applied after mechanical fragmentation and assess these effects radiographically in bone healing in dogs subjected to tibial plateau levelling osteotomy (TPLO). Twenty dogs with unilateral cranial cruciate ligament disease were enrolled and randomly assigned to the treatment group (MFAT) or the control group (NT). The MFAT group underwent TPLO and autologous MFAT intra-articular administration, while the NT group underwent TPLO alone. Adipose tissue was collected from the thigh region, and MFAT was obtained by mechanical fragmentation at the end of the surgery. The patients were subjected to X-ray examination preoperatively, immediately postoperatively (T0), and at 4 (T1) and 8 (T2) weeks postoperatively. Two radiographic scores that had previously been described for the evaluation of bone healing after TPLO were used. A 12-point scoring system (from 0 = no healing to 12 = complete remodelling) was used at T0, T1, and T2, while a 5-point scoring system (from 0 = no healing to 4 = 76-100% of healing) was used at T1 and T2. The median healing scores were significantly higher at T1 and T2 for the MFAT group compared with the NT group for the 12-point (p < 0.05) and 5-point (p < 0.05) scoring systems. The intra-articular injection of autologous microfragmented adipose tissue can accelerate bone healing after TPLO without complications.
Collapse
Affiliation(s)
- Luca Pennasilico
- School of Bioscience and Veterinary Medicine, University of Camerino, 62024 Matelica, Italy
| | - Caterina Di Bella
- School of Bioscience and Veterinary Medicine, University of Camerino, 62024 Matelica, Italy
| | - Sara Sassaroli
- School of Bioscience and Veterinary Medicine, University of Camerino, 62024 Matelica, Italy
| | | | | | | |
Collapse
|
43
|
Majood M, Selvam A, Agrawal O, Chaurasia R, Rawat S, Mohanty S, Mukherjee M. Biogenic Carbon Quantum Dots as a Neoteric Inducer in the Game of Directing Chondrogenesis. ACS APPLIED MATERIALS & INTERFACES 2023; 15:19997-20011. [PMID: 37042793 DOI: 10.1021/acsami.3c02007] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
The journey into the field of stem cell biology has been an endeavor of paramount advancement in biomedicine, establishing new horizons in the avenue of materiobiology. The creative drive of the scientific community focuses on ameliorating the utilization of stem cells, which is currently untapped on a large scale. With similar motivation, we present a nascent strategy of maneuvering biogenic carbon quantum dots (CQDs) to eclipse the toxic hurdles of chemical synthesis of carbon allotropes to serve as a biocompatible trident in stem cell biology employing a three-prong action of stem cell differentiation, imaging, and migration. The derivation of CQDs from garlic peels as a biogenic precursor abets in realizing the optophysical features of CQDs to image mesenchymal stem cells without hampering the biological systems with cytotoxicity. We report the versatility of biogenic CQDs to generate reactive oxygen species (ROS) to robustly influence stem cell migration and concomitantly chondrocyte differentiation from human Wharton's jelly mesenchymal stem cells (hWJ-MSCs). This was orchestrated without the use of chondrogenic induction factors, which was confirmed from the expression of chondrogenic markers (Col II, Col X, ACAN). Even the collagen content of cells incubated with CQDs was quite comparable with that of chondrocyte-induced cells. Thus, we empirically propose garlic peel-derived CQDs as a tangible advancement in stem cell biology from a materiobiological frame of reference to hone significant development in this arena.
Collapse
Affiliation(s)
- Misba Majood
- Amity Institute of Click Chemistry Research and Studies, Amity University, Noida, Uttar Pradesh 201303, India
| | - Abhyavartin Selvam
- Amity Institute of Click Chemistry Research and Studies, Amity University, Noida, Uttar Pradesh 201303, India
- Amity Institute of Nanotechnology, Amity University, Noida, Uttar Pradesh 201303, India
| | - Omnarayan Agrawal
- Amity Institute of Click Chemistry Research and Studies, Amity University, Noida, Uttar Pradesh 201303, India
| | - Radhika Chaurasia
- Amity Institute of Click Chemistry Research and Studies, Amity University, Noida, Uttar Pradesh 201303, India
| | - Sonali Rawat
- Stem Cells Facility, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Sujata Mohanty
- Stem Cells Facility, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Monalisa Mukherjee
- Amity Institute of Click Chemistry Research and Studies, Amity University, Noida, Uttar Pradesh 201303, India
| |
Collapse
|
44
|
Prakash N, Kim J, Jeon J, Kim S, Arai Y, Bello AB, Park H, Lee SH. Progress and emerging techniques for biomaterial-based derivation of mesenchymal stem cells (MSCs) from pluripotent stem cells (PSCs). Biomater Res 2023; 27:31. [PMID: 37072836 PMCID: PMC10114339 DOI: 10.1186/s40824-023-00371-0] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 03/26/2023] [Indexed: 04/20/2023] Open
Abstract
The use of mesenchymal stem cells (MSCs) for clinical purposes has skyrocketed in the past decade. Their multilineage differentiation potentials and immunomodulatory properties have facilitated the discovery of therapies for various illnesses. MSCs can be isolated from infant and adult tissue sources, which means they are easily available. However, this raises concerns because of the heterogeneity among the various MSC sources, which limits their effective use. Variabilities arise from donor- and tissue-specific differences, such as age, sex, and tissue source. Moreover, adult-sourced MSCs have limited proliferation potentials, which hinders their long-term therapeutic efficacy. These limitations of adult MSCs have prompted researchers to develop a new method for generating MSCs. Pluripotent stem cells (PSCs), such as embryonic stem cells and induced PSCs (iPSCs), can differentiate into various types of cells. Herein, a thorough review of the characteristics, functions, and clinical importance of MSCs is presented. The existing sources of MSCs, including adult- and infant-based sources, are compared. The most recent techniques for deriving MSCs from iPSCs, with a focus on biomaterial-assisted methods in both two- and three-dimensional culture systems, are listed and elaborated. Finally, several opportunities to develop improved methods for efficiently producing MSCs with the aim of advancing their various clinical applications are described.
Collapse
Affiliation(s)
- Nityanand Prakash
- Department of Biomedical Engineering, Dongguk University, Seoul, 04620, Korea
| | - Jiseong Kim
- Department of Biomedical Engineering, Dongguk University, Seoul, 04620, Korea
| | - Jieun Jeon
- Department of Biomedical Engineering, Dongguk University, Seoul, 04620, Korea
| | - Siyeon Kim
- Department of Biomedical Engineering, Dongguk University, Seoul, 04620, Korea
| | - Yoshie Arai
- Department of Biomedical Engineering, Dongguk University, Seoul, 04620, Korea
| | - Alvin Bacero Bello
- Department of Biomedical Engineering, Dongguk University, Seoul, 04620, Korea.
| | - Hansoo Park
- School of Integrative Engineering, Chung-Ang University, Seoul, 06911, Korea.
| | - Soo-Hong Lee
- Department of Biomedical Engineering, Dongguk University, Seoul, 04620, Korea.
| |
Collapse
|
45
|
Berni M, Brancato AM, Torriani C, Bina V, Annunziata S, Cornella E, Trucchi M, Jannelli E, Mosconi M, Gastaldi G, Caliogna L, Grassi FA, Pasta G. The Role of Low-Level Laser Therapy in Bone Healing: Systematic Review. Int J Mol Sci 2023; 24:7094. [PMID: 37108257 PMCID: PMC10139216 DOI: 10.3390/ijms24087094] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 04/06/2023] [Accepted: 04/10/2023] [Indexed: 04/29/2023] Open
Abstract
Low-level laser therapy (LLLT) is a treatment that is increasingly used in orthopedics practices. In vivo and in vitro studies have shown that low-level laser therapy (LLLT) promotes angiogenesis, fracture healing and osteogenic differentiation of stem cells. However, the underlying mechanisms during bone formation remain largely unknown. Factors such as wavelength, energy density, irradiation and frequency of LLLT can influence the cellular mechanisms. Moreover, the effects of LLLT are different according to cell types treated. This review aims to summarize the current knowledge of the molecular pathways activated by LLLT and its effects on the bone healing process. A better understanding of the cellular mechanisms activated by LLLT can improve its clinical application.
Collapse
Affiliation(s)
- Micaela Berni
- Orthopedics and Traumatology Clinic, IRCCS Policlinico San Matteo Foundation, 27100 Pavia, Italy
| | - Alice Maria Brancato
- Orthopedics and Traumatology Clinic, IRCCS Policlinico San Matteo Foundation, 27100 Pavia, Italy
| | - Camilla Torriani
- Orthopedics and Traumatology Clinic, IRCCS Policlinico San Matteo Foundation, 27100 Pavia, Italy
| | - Valentina Bina
- Department of Molecular Medicine, University of Pavia, 27100 Pavia, Italy
| | - Salvatore Annunziata
- Orthopedics and Traumatology Clinic, IRCCS Policlinico San Matteo Foundation, 27100 Pavia, Italy
| | - Elena Cornella
- Orthopedics and Traumatology Clinic, IRCCS Policlinico San Matteo Foundation, 27100 Pavia, Italy
| | - Michelangelo Trucchi
- Orthopedics and Traumatology Clinic, IRCCS Policlinico San Matteo Foundation, 27100 Pavia, Italy
| | - Eugenio Jannelli
- Orthopedics and Traumatology Clinic, IRCCS Policlinico San Matteo Foundation, 27100 Pavia, Italy
| | - Mario Mosconi
- Orthopedics and Traumatology Clinic, IRCCS Policlinico San Matteo Foundation, 27100 Pavia, Italy
| | - Giulia Gastaldi
- Department of Molecular Medicine, University of Pavia, 27100 Pavia, Italy
- Centre for Health Technologies, University of Pavia, 27100 Pavia, Italy
| | - Laura Caliogna
- Orthopedics and Traumatology Clinic, IRCCS Policlinico San Matteo Foundation, 27100 Pavia, Italy
| | - Federico Alberto Grassi
- Orthopedics and Traumatology Clinic, IRCCS Policlinico San Matteo Foundation, 27100 Pavia, Italy
- Centre for Health Technologies, University of Pavia, 27100 Pavia, Italy
- Department of Clinical, Surgical, Diagnostic and Pediatric Sciences, University of Pavia, 27100 Pavia, Italy
| | - Gianluigi Pasta
- Orthopedics and Traumatology Clinic, IRCCS Policlinico San Matteo Foundation, 27100 Pavia, Italy
| |
Collapse
|
46
|
Frost K, Naylor AJ, McGettrick HM. The Ying and Yang of Sphingosine-1-Phosphate Signalling within the Bone. Int J Mol Sci 2023; 24:6935. [PMID: 37108099 PMCID: PMC10139073 DOI: 10.3390/ijms24086935] [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: 02/06/2023] [Revised: 03/28/2023] [Accepted: 04/04/2023] [Indexed: 04/29/2023] Open
Abstract
Bone remodelling is a highly active and dynamic process that involves the tight regulation of osteoblasts, osteoclasts, and their progenitors to allow for a balance of bone resorption and formation to be maintained. Ageing and inflammation are risk factors for the dysregulation of bone remodelling. Once the balance between bone formation and resorption is lost, bone mass becomes compromised, resulting in disorders such as osteoporosis and Paget's disease. Key molecules in the sphingosine-1-phosphate signalling pathway have been identified for their role in regulating bone remodelling, in addition to its more recognised role in inflammatory responses. This review discusses the accumulating evidence for the different, and, in certain circumstances, opposing, roles of S1P in bone homeostasis and disease, including osteoporosis, Paget's disease, and inflammatory bone loss. Specifically, we describe the current, often conflicting, evidence surrounding S1P function in osteoblasts, osteoclasts, and their precursors in health and disease, concluding that S1P may be an effective biomarker of bone disease and also an attractive therapeutic target for disease.
Collapse
Affiliation(s)
| | - Amy J. Naylor
- Rheumatology Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham B15 2TT, UK
| | | |
Collapse
|
47
|
Sadek AA, Abd-Elkareem M, Abdelhamid HN, Moustafa S, Hussein K. Repair of critical-sized bone defects in rabbit femurs using graphitic carbon nitride (g-C 3N 4) and graphene oxide (GO) nanomaterials. Sci Rep 2023; 13:5404. [PMID: 37012344 PMCID: PMC10070441 DOI: 10.1038/s41598-023-32487-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 03/28/2023] [Indexed: 04/05/2023] Open
Abstract
Various biomaterials have been evaluated to enhance bone formation in critical-sized bone defects; however, the ideal scaffold is still missing. The objective of this study was to investigate the in vitro and in vivo regenerative capacity of graphitic carbon nitride (g-C3N4) and graphene oxide (GO) nanomaterials to stimulate critical-sized bone defect regeneration. The in vitro cytotoxicity and hemocompatibility of g-C3N4 and GO were evaluated, and their potential to induce the in vitro osteogenesis of human fetal osteoblast (hFOB) cells was assessed using qPCR. Then, bone defect in femoral condyles was created in rabbits and left empty as control or filled with either g-C3N4 or GO. The osteogenesis of the different implanted scaffolds was evaluated after 4, 8, and 12 weeks of surgery using X-ray, computed tomography (CT), macro/microscopic examinations, and qPCR analysis of osteocalcin (OC) and osteopontin (OP) expressions. Both materials displayed good cell viability and hemocompatibility with enhanced collagen type-I (Col-I), OC, and OP expressions of the hFOB cells. Compared to the control group, the bone healing process in g-C3N4 and GO groups was promoted in vivo. Moreover, complete healing of the bone defect was observed radiologically and grossly in g-C3N4 implanted group. Additionally, g-C3N4 implanted group showed higher percentages of osteoid tissue, mature collagen, biodegradation, and expressions of OC and OP. In conclusion, our results revealed that g-C3N4 and GO nanomaterials could induce osteogenesis in critical-sized bone defects.
Collapse
Affiliation(s)
- Ahmed Abdelrahiem Sadek
- Department of Surgery, Anesthesiology and Radiology, Faculty of Veterinary Medicine, Assiut University, Assiut, 71526, Egypt
| | - Mahmoud Abd-Elkareem
- Department of Cell and Tissues, Faculty of Veterinary Medicine, Assiut University, Assiut, Egypt
| | - Hani Nasser Abdelhamid
- Advanced Multifunctional Materials Laboratory, Department of Chemistry, Faculty of Science, Assiut University, Assiut, Egypt
- Proteomics Laboratory for Clinical Research and Materials Science, Department of Chemistry, Faculty of Science, Assiut University, Assiut, Egypt
- Nanotechnology Research Centre (NTRC), The British University in Egypt (BUE), Suez Desert Road, El-Sherouk City, 11837, Cairo, Egypt
| | - Samia Moustafa
- Department of Surgery, Anesthesiology and Radiology, Faculty of Veterinary Medicine, Assiut University, Assiut, 71526, Egypt
| | - Kamal Hussein
- Department of Surgery, Anesthesiology and Radiology, Faculty of Veterinary Medicine, Assiut University, Assiut, 71526, Egypt.
| |
Collapse
|
48
|
Hong Z, Zhang Y, Chen J, Bi Q. Adipose-derived stromal vascular fraction injection following core decompression and biochemistry artificial bone graft implantation in osteonecrosis of the femoral head. INTERNATIONAL ORTHOPAEDICS 2023; 47:1481-1486. [PMID: 36991282 DOI: 10.1007/s00264-023-05792-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 03/21/2023] [Indexed: 03/31/2023]
Abstract
PURPOSE To determine how adipose-derived stromal vascular fraction (SVF) injection following core decompression (CD) and biochemistry artificial bone graft implantation affects outcomes in patients with osteonecrosis of the femoral head (ONFH). METHODS A total of 19 patients (28 hips) with stage I-IIIA ONFH received adipose-derived SVF injection and combined core decompression and biochemistry artificial bone graft implantation, followed up for a minimum of two years. Disease progression was evaluated according to the Association Research Circulation Osseous (ARCO) staging system, and the change of the ratio of the necrotic volume to femoral head volume was calculated with MRI before and after operation. RESULTS At the last follow-up, 15 hips remained stable, and 13 hips had a progression, according to the ARCO staging system. A total of eight hips (5 with ARCO stage II and 3 with staged IIIA at baseline) progressed to post-collapse stage (stage IIIB-IV). In total, seven of eight hips with post-collapse stage and one with IIIA stage at follow-up converted to THAs in an average of 17.5 months (range, 11-68 months) postoperatively. The mean ratio of the necrotic lesion volume to the femoral head significantly decreased in hips with ARCO stage I (17.9 ± 3.0% to 9.8 ± 1.3%, p = 0.012, Δ necrosis ratio = 8.1 ± 4.2%) and stage II (22.7 ± 6.3% to 17.1 ± 9.4%, p = 0.001, Δ necrosis ratio = 5.7 ± 6.6%) at baseline. For the eight hips that progressed to post-collapse stage, the mean necrosis ratio increased from 27.4 ± 5.4% to 31.1 ± 4.0% (p = 0.146), Δ necrosis ratio = - 3.7 ± 3.9%. For the other 20 hips radiological survived, the mean necrosis ratio improved from 19.9 ± 4.4% to 11.8 ± 3.3% (p < 0.001), with Δ necrosis ratio = 8.1 ± 4.9%. CONCLUSION Adipose-derived SVF injection following core decompression and biochemistry artificial bone graft implantation is safe and could effectively repair the necrosis lesion and delay disease progression in patients with early-stage ONFH.
Collapse
Affiliation(s)
- Zheping Hong
- Suzhou Medical College of Soochow University, Suzhou, Jiangsu, China
- Department of Orthopaedic Surgery, Zhejiang Provincial People's Hospital and People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Yin Zhang
- Department of Orthopaedic Surgery, Zhejiang Provincial People's Hospital and People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Jihang Chen
- Department of Orthopaedic Surgery, Zhejiang Provincial People's Hospital and People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Qing Bi
- Suzhou Medical College of Soochow University, Suzhou, Jiangsu, China.
- Department of Orthopaedic Surgery, Zhejiang Provincial People's Hospital and People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang, China.
| |
Collapse
|
49
|
Feng Z, Jin M, Liang J, Kang J, Yang H, Guo S, Sun X. Insight into the effect of biomaterials on osteogenic differentiation of mesenchymal stem cells: A review from a mitochondrial perspective. Acta Biomater 2023; 164:1-14. [PMID: 36972808 DOI: 10.1016/j.actbio.2023.03.032] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 03/02/2023] [Accepted: 03/21/2023] [Indexed: 03/29/2023]
Abstract
Bone damage may be triggered by a variety of factors, and the damaged area often requires a bone graft. Bone tissue engineering can serve as an alternative strategy for repairing large bone defects. Mesenchymal stem cells (MSCs), the progenitor cells of connective tissue, have become an important tool for tissue engineering due to their ability to differentiate into a variety of cell types. The precise regulation of the growth and differentiation of the stem cells used for bone regeneration significantly affects the efficiency of this type of tissue engineering. During the process of osteogenic induction, the dynamics and function of localized mitochondria are altered. These changes may also alter the microenvironment of the therapeutic stem cells and result in mitochondria transfer. Mitochondrial regulation not only affects the induction/rate of differentiation, but also influences its direction, determining the final identity of the differentiated cell. To date, bone tissue engineering research has mainly focused on the influence of biomaterials on phenotype and nuclear genotype, with few studies investigating the role of mitochondria. In this review, we provide a comprehensive summary of researches into the role of mitochondria in MSCs differentiation and critical analysis regarding smart biomaterials that are able to "programme" mitochondria modulation was proposed. STATEMENT OF SIGNIFICANCE: : • This review proposed the precise regulation of the growth and differentiation of the stem cells used to seed bone regeneration. • This review addressed the dynamics and function of localized mitochondria during the process of osteogenic induction and the effect of mitochondria on the microenvironment of stem cells. • This review summarized biomaterials which affect the induction/rate of differentiation, but also influences its direction, determining the final identity of the differentiated cell through the regulation of mitochondria.
Collapse
Affiliation(s)
- Ziyi Feng
- Department of Plastic Surgery, The First Hospital of China Medical University, No. 155, Nanjing North Street, Heping District, Shenyang, 110002 Liaoning Province, China
| | - Meiqi Jin
- School of Intelligent Medicine, China Medical University, No.77, Puhe Road, Shenyang, 110122, Liaoning Province, China
| | - Junzhi Liang
- Center of Reproductive Medicine, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping, Shenyang, 110004 Liaoning Province, China
| | - Junning Kang
- Department of Neurosurgery, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping, Shenyang, 110004 Liaoning Province, China
| | - Huazhe Yang
- School of Intelligent Medicine, China Medical University, No.77, Puhe Road, Shenyang, 110122, Liaoning Province, China.
| | - Shu Guo
- Department of Plastic Surgery, The First Hospital of China Medical University, No. 155, Nanjing North Street, Heping District, Shenyang, 110002 Liaoning Province, China.
| | - Xiaoting Sun
- School of Forensic Medicine, China Medical University, No.77, Puhe Road, Shenyang, 110122, Liaoning Province, China.
| |
Collapse
|
50
|
Liu J, Yang L, Liu K, Gao F. Hydrogel scaffolds in bone regeneration: Their promising roles in angiogenesis. Front Pharmacol 2023; 14:1050954. [PMID: 36860296 PMCID: PMC9968752 DOI: 10.3389/fphar.2023.1050954] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Accepted: 02/03/2023] [Indexed: 02/16/2023] Open
Abstract
Bone tissue engineering (BTE) has become a hopeful potential treatment strategy for large bone defects, including bone tumors, trauma, and extensive fractures, where the self-healing property of bone cannot repair the defect. Bone tissue engineering is composed of three main elements: progenitor/stem cells, scaffold, and growth factors/biochemical cues. Among the various biomaterial scaffolds, hydrogels are broadly used in bone tissue engineering owing to their biocompatibility, controllable mechanical characteristics, osteoconductive, and osteoinductive properties. During bone tissue engineering, angiogenesis plays a central role in the failure or success of bone reconstruction via discarding wastes and providing oxygen, minerals, nutrients, and growth factors to the injured microenvironment. This review presents an overview of bone tissue engineering and its requirements, hydrogel structure and characterization, the applications of hydrogels in bone regeneration, and the promising roles of hydrogels in bone angiogenesis during bone tissue engineering.
Collapse
Affiliation(s)
- Jun Liu
- Department of Hand Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Lili Yang
- Department of Spinal Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Kexin Liu
- Department of Gastrointestinal Colorectal Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Feng Gao
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, China,*Correspondence: Feng Gao,
| |
Collapse
|