1
|
Lu X, Zhao Y, Peng X, Lu C, Wu Z, Xu H, Qin Y, Xu Y, Wang Q, Hao Y, Geng D. Comprehensive Overview of Interface Strategies in Implant Osseointegration. ADVANCED FUNCTIONAL MATERIALS 2024. [DOI: 10.1002/adfm.202418849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Indexed: 01/05/2025]
Abstract
AbstractWith the improvement of implant design and the expansion of application scenarios, orthopedic implants have become a common surgical option for treating fractures and end‐stage osteoarthritis. Their common goal is rapidly forming and long‐term stable osseointegration. However, this fixation effect is limited by implant surface characteristics and peri‐implant bone tissue activity. Therefore, this review summarizes the strategies of interface engineering (osteogenic peptides, growth factors, and metal ions) and treatment methods (porous nanotubes, hydrogel embedding, and other load‐release systems) through research on its biological mechanism, paving the way to achieve the adaptation of both and coordination between different strategies. With the transition of the osseointegration stage, interface engineering strategies have demonstrated varying therapeutic effects. Especially, the activity of osteoblasts runs almost through the entire process of osseointegration, and their physiological activities play a dominant role in bone formation. Furthermore, diseases impacting bone metabolism exacerbate the difficulty of achieving osseointegration. This review aims to assist future research on osseointegration engineering strategies to improve implant‐bone fixation, promote fracture healing, and enhance post‐implantation recovery.
Collapse
Affiliation(s)
- Xiaoheng Lu
- Department of Orthopedics The First Affiliated Hospital of Soochow University 188 Shizi Street Suzhou Jiangsu 215006 China
| | - Yuhu Zhao
- Department of Orthopedics The First Affiliated Hospital of Soochow University 188 Shizi Street Suzhou Jiangsu 215006 China
| | - Xiaole Peng
- Department of Orthopedics The First Affiliated Hospital of Soochow University 188 Shizi Street Suzhou Jiangsu 215006 China
- Department of Orthopedics The First Affiliated Hospital of Chongqing Medical University 1 Youyi Street Chongqing 400016 China
| | - Chengyao Lu
- Department of Orthopedics The First Affiliated Hospital of Soochow University 188 Shizi Street Suzhou Jiangsu 215006 China
| | - Zebin Wu
- Department of Orthopedics The First Affiliated Hospital of Soochow University 188 Shizi Street Suzhou Jiangsu 215006 China
| | - Hao Xu
- Department of Orthopedics The First Affiliated Hospital of Soochow University 188 Shizi Street Suzhou Jiangsu 215006 China
| | - Yi Qin
- Department of Orthopedics The First Affiliated Hospital of Soochow University 188 Shizi Street Suzhou Jiangsu 215006 China
| | - Yaozeng Xu
- Department of Orthopedics The First Affiliated Hospital of Soochow University 188 Shizi Street Suzhou Jiangsu 215006 China
| | - Qing Wang
- Department of Orthopedics The First Affiliated Hospital of Soochow University 188 Shizi Street Suzhou Jiangsu 215006 China
| | - Yuefeng Hao
- Orthopedics and Sports Medicine Center The Affiliated Suzhou Hospital of Nanjing Medical University 242 Guangji Street Suzhou Jiangsu 215006 China
| | - Dechun Geng
- Department of Orthopedics The First Affiliated Hospital of Soochow University 188 Shizi Street Suzhou Jiangsu 215006 China
| |
Collapse
|
2
|
Qu Z, Zhao S, Zhang Y, Wang X, Yan L. Natural Compounds for Bone Remodeling: Targeting osteoblasts and relevant signaling pathways. Biomed Pharmacother 2024; 180:117490. [PMID: 39332184 DOI: 10.1016/j.biopha.2024.117490] [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: 06/23/2024] [Revised: 09/10/2024] [Accepted: 09/20/2024] [Indexed: 09/29/2024] Open
Abstract
In the process of bone metabolism and bone remodeling, bone marrow mesenchymal stem cells (BM-MSCs) differentiate into osteoblasts (OBs) under certain conditions to enable the formation of new bone, and normal bone reconstruction and pathological bone alteration are closely related to the differentiation and proliferation functions of OBs. Osteogenic differentiation of BM-MSCs involves multiple signaling pathways, which function individually but interconnect intricately to form a complex signaling regulatory network. Natural compounds have fewer adverse effects than chemically synthesized drugs, optimize bone health, and are more suitable for long-term use. In this paper, we focus on OBs, summarize the current research progress of signaling pathways related to OBs differentiation, and review the molecular mechanisms by which chemically synthesized drugs with potential anti-osteoporosis properties regulate OBs-mediated bone formation.
Collapse
Affiliation(s)
- Zechao Qu
- Department of Spinal Surgery, Honghui Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Songchuan Zhao
- Department of Spinal Surgery, Honghui Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yong Zhang
- Department of Spinal Surgery, Honghui Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xiaohao Wang
- Department of Spinal Surgery, Honghui Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Liang Yan
- Department of Spinal Surgery, Honghui Hospital of Xi'an Jiaotong University, Xi'an, China.
| |
Collapse
|
3
|
Zhu S, Chen W, Masson A, Li YP. Cell signaling and transcriptional regulation of osteoblast lineage commitment, differentiation, bone formation, and homeostasis. Cell Discov 2024; 10:71. [PMID: 38956429 PMCID: PMC11219878 DOI: 10.1038/s41421-024-00689-6] [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: 07/07/2023] [Accepted: 05/04/2024] [Indexed: 07/04/2024] Open
Abstract
The initiation of osteogenesis primarily occurs as mesenchymal stem cells undergo differentiation into osteoblasts. This differentiation process plays a crucial role in bone formation and homeostasis and is regulated by two intricate processes: cell signal transduction and transcriptional gene expression. Various essential cell signaling pathways, including Wnt, BMP, TGF-β, Hedgehog, PTH, FGF, Ephrin, Notch, Hippo, and Piezo1/2, play a critical role in facilitating osteoblast differentiation, bone formation, and bone homeostasis. Key transcriptional factors in this differentiation process include Runx2, Cbfβ, Runx1, Osterix, ATF4, SATB2, and TAZ/YAP. Furthermore, a diverse array of epigenetic factors also plays critical roles in osteoblast differentiation, bone formation, and homeostasis at the transcriptional level. This review provides an overview of the latest developments and current comprehension concerning the pathways of cell signaling, regulation of hormones, and transcriptional regulation of genes involved in the commitment and differentiation of osteoblast lineage, as well as in bone formation and maintenance of homeostasis. The paper also reviews epigenetic regulation of osteoblast differentiation via mechanisms, such as histone and DNA modifications. Additionally, we summarize the latest developments in osteoblast biology spurred by recent advancements in various modern technologies and bioinformatics. By synthesizing these insights into a comprehensive understanding of osteoblast differentiation, this review provides further clarification of the mechanisms underlying osteoblast lineage commitment, differentiation, and bone formation, and highlights potential new therapeutic applications for the treatment of bone diseases.
Collapse
Affiliation(s)
- Siyu Zhu
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane University, New Orleans, LA, USA
| | - Wei Chen
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane University, New Orleans, LA, USA.
| | - Alasdair Masson
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane University, New Orleans, LA, USA
| | - Yi-Ping Li
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane University, New Orleans, LA, USA.
| |
Collapse
|
4
|
Wang B, Wang W, Li J, Li J. Zinc finger protein 36 like 2-histone deacetylase 1 axis is involved in the bone responses to mechanical stress. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167162. [PMID: 38604490 DOI: 10.1016/j.bbadis.2024.167162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 03/26/2024] [Accepted: 04/04/2024] [Indexed: 04/13/2024]
Abstract
The molecular mechanism underlying the promotion of fracture healing by mechanical stimuli remains unclear. The present study aimed to investigate the role of zinc finger protein 36 like 2 (ZFP36L2)-histone deacetylase 1 (HDAC1) axis on the osteogenic responses to moderate mechanical stimulation. Appropriate stimulation of fluid shear stress (FSS) was performed on MC3T3-E1 cells transduced with ZFP36L2 and HDAC1 recombinant adenoviruses, aiming to validate the influence of mechanical stress on the expression of ZFP36L2-HDAC1 and the osteogenic differentiation and mineralization. The results showed that moderate FSS stimulation significantly upregulated the expression of ZFP36L2 in MC3T3-E1 cells (p < 0.01). The overexpression of ZFP36L1 markedly enhanced the levels of osteogenic differentiation markers, including bone morphogenetic protein 2 (BMP2), runt-related transcription factor 2 (RUNX2), alkaline phosphatase (ALP), Osterix, and collagen type I alpha 1 (COL1A1) (p < 0.01). ZFP36L2 accelerated the degradation of HDAC1 by specifically binding to its 3' UTR region, thereby fulfilling its function at the post-transcriptional regulatory gene level and promoting the osteogenic differentiation and mineralization fate of cells. Mechanical unloading notably diminished/elevated the expression of ZFP36L2/HDAC1, decreased bone mineral density and bone volume fraction, hindered the release of osteogenic-related factors and vascular endothelial growth factor in callus tissue (p < 0.01), and was detrimental to fracture healing. Collectively, proper stress stimulation plays a crucial role in facilitating osteogenesis through the promotion of ZFP36L2 and subsequent degradation of HDAC1. Targeting ZFP36L2-HDAC1 axis may provide promising insights to enhance bone defect healing.
Collapse
Affiliation(s)
- Bin Wang
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang 110004, People's Republic of China
| | - Wei Wang
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang 110004, People's Republic of China
| | - Jingyu Li
- Department of Ultrasound, Shengjing Hospital of China Medical University, Shenyang 110004, People's Republic of China.
| | - Jianjun Li
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang 110004, People's Republic of China.
| |
Collapse
|
5
|
Bao DY, Yang Y, Tong X, Qin HY. Activation of wnt/β-catenin signaling pathway down regulated osteogenic differentiation of bone marrow-derived stem cells in an anhidrotic ectodermal dysplasia patient with EDA/EDAR/EDARADD mutation. Heliyon 2024; 10:e23057. [PMID: 38169761 PMCID: PMC10758735 DOI: 10.1016/j.heliyon.2023.e23057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 10/29/2023] [Accepted: 11/24/2023] [Indexed: 01/05/2024] Open
Abstract
Objective To explore the mechanism by which the Wnt/β-catenin pathway induces osteogenic differentiation of bone marrow-derived stem cells (BMSCs) in anhidrotic ectodermal dysplasia (AED) with an Ectodysplasin A (EDA)/EDA receptor (EDAR)/EDARADD mutation. Methods An AED patient served as the AED group, whereas the other patients without AED were included in the normal group. Peripheral venous blood collected from the AED patient was subjected to whole-genome resequencing. BMSCs from the mandible of patients with AED and normal individuals were isolated and cultured in vitro. Cell proliferation assay was performed to compare the growth speed of BMSCs between the AED and normal groups. CHIR-99021, an activator of the Wnt/β-catenin pathway and XAV-939, an inhibitor, was used to manage BMSCs in an osteogenic environment in both groups. The expression of β-catenin was detected by quantitative polymerase chain reaction, while that of RUNX2 was detected by western blotting. Alizarin red was used for staining. Results A novel mutation (c.152T > A in EDA) and two known mutations (c.1109T > C in EDAR and c.27G > A in EDARADD) were identified. The growth rate in the normal group was higher than that in the AED group. In the normal group, the number and size of calcified nodes and the expression of RUNX-2 increased with CHIR-99021 treatment, which could be inhibited by XAV-939. In contrast, CHIR-99021 inhibited osteogenesis in the AED group and this effect was promoted by XAV-939. Conclusion Activation of the Wnt/β-catenin pathway downregulates osteogenesis of BMSCs in AED patients with EDA/EDAR/EDARADD gene mutations. Further investigation in more AED patients is required, given the wide range of mutations involved in AED.
Collapse
Affiliation(s)
- Dong-yu Bao
- Department of Stomatology, the Affiliated Drum Tower Hospital of Nanjing University Medical School. 321 Zhongshan Road, Nanjing, 210008, China
- Department of Dental Implantology, Nanjing Stomatological Hospital, Medical School of Nanjing University, No.30 Zhongyang Road, Nanjing, 210008, China
| | - Yun Yang
- Department of Stomatology, the Affiliated Drum Tower Hospital of Nanjing University Medical School. 321 Zhongshan Road, Nanjing, 210008, China
| | - Xin Tong
- Department of Dental Implantology, Nanjing Stomatological Hospital, Medical School of Nanjing University, No.30 Zhongyang Road, Nanjing, 210008, China
| | - Hai-yan Qin
- Department of Stomatology, the Affiliated Drum Tower Hospital of Nanjing University Medical School. 321 Zhongshan Road, Nanjing, 210008, China
| |
Collapse
|
6
|
Zhang M, Xu B, Li W, Yu B, Peng H, Gui F, Ai F, Chen Z. lncRNA CCAT2 Protects Against Cardiomyocyte Injury After Myocardial Ischemia/Reperfusion by Regulating BMI1 Expression. Int Heart J 2024; 65:279-291. [PMID: 38556336 DOI: 10.1536/ihj.23-569] [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] [Indexed: 04/02/2024]
Abstract
Myocardial ischemia/reperfusion (I/R) decreases cardiac function and efficiency. Accumulating evidence suggests that long noncoding RNAs (lncRNAs) have been linked to the cellular processes of myocardial I/R injury. The present investigation elucidated the function of lncRNA colon cancer-associated transcript 2 (CCAT2) in myocardial I/R injury and the related mechanisms.AC16 cardiomyocytes were exposed to hypoxia (16 hours) /reoxygenation (6 hours) (H/R) to mimic myocardial I/R models in vitro. CCAT2 and microRNA (miR) -539-3p expressions in AC16 cardiomyocytes were measured using real-time quantitative polymerase chain reaction. B-cell-specific Moloney murine leukemia virus insertion region 1 (BMI1) protein levels in AC16 cardiomyocytes were determined by western blotting. Cell viability, lactate dehydrogenase (LDH) leakage, reactive oxygen species (ROS) levels, mitochondrial membrane potential, and apoptosis were detected using Counting Kit-8, LDH Assay Kit, dihydroethidium assay, 5,5',6,6'-tetrachloro1,1',3,3'-tetramethylbenzimidazolylcarbocyanine iodide staining, flow cytometry, and western blotting, respectively. The interactions between the molecules were confirmed using the dual-luciferase gene reporter. The wingless/integrated/beta-catenin (Wnt/β-catenin) pathway under the H/R condition was detected by western blotting.CCAT2 and BMI1 mRNA expressions were reduced in H/R-exposed AC16 cardiomyocytes. CCAT2 overexpression exerted protective effects against H/R-induced cardiomyocyte injury, as demonstrated by increased cell viability and mitochondrial membrane potential and decreased LDH leakage, ROS levels, and apoptosis. In addition, CCAT2 positively regulated BMI1 expression by binding to miR-539-3p. CCAT2 knockdown or miR-539-3p overexpression restrained the protective effects of BMI1 against H/R-induced cardiomyocyte injury. In addition, miR-539-3p overexpression reversed the protective effects of CCAT2. Furthermore, CCAT2 activated the Wnt/β-catenin pathway under the H/R condition via the miR-539-3p/BMI1 axis.Overall, this investigation showed the protective effects of the CCAT2/miR-539-3p/BMI1/Wnt/β-catenin regulatory axis against cardiomyocyte injury induced by H/R.
Collapse
Affiliation(s)
- Mengli Zhang
- Department of Emergency, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology
| | - Bei Xu
- Department of Cardiovasology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology
| | - Wei Li
- Department of Emergency, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology
| | - Bo Yu
- Department of Emergency, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology
| | - Huan Peng
- Department of Emergency, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology
| | - Feng Gui
- Department of Emergency, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology
| | - Fen Ai
- Department of Emergency, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology
| | - Zhen Chen
- Department of Emergency, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology
| |
Collapse
|
7
|
Zhou G, Zhang X, Gu Z, Zhao J, Luo M, Liu J. Research progress on the treatment of knee osteoarthritis combined with osteoporosis by single-herb Chinese medicine and compound. Front Med (Lausanne) 2023; 10:1254086. [PMID: 37841009 PMCID: PMC10568449 DOI: 10.3389/fmed.2023.1254086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 09/11/2023] [Indexed: 10/17/2023] Open
Abstract
Knee osteoarthritis (KOA) is a degenerative disease with synovial inflammation, articular surface cartilage degeneration, meniscus degeneration, ligament and muscle changes, subchondral bone changes, and osteophyte formation around the joint as the main pathological changes. Osteoporosis (OP) is a disease characterized by low bone mass and deterioration of the microstructure of bone tissue. KOA and OP are both geriatric diseases, and the incidence of KOA combined with OP is high, but there is a lack of specific drugs, and the major treatments are limited to drug therapy. Most traditional Chinese medicine (TCM) treatments use plant-based natural products, and they help patients obtain good clinical benefits and at the same time provide researchers with ideas to study the mechanism of disease occurrence and the relationship between the two diseases. This article summarizes the research progress of TCM monomers and TCM compounds that are frequently used to treat KOA combined with OP to provide ideas for future clinical treatments and related basic research.
Collapse
Affiliation(s)
- Guanghui Zhou
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xianquan Zhang
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhuoxu Gu
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jinlong Zhao
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Minghui Luo
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jun Liu
- Bone and Joint Research Team of Degeneration and Injury, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, China
- Guangdong Second Traditional Chinese Medicine Hospital, Guangdong Province Engineering Technology Research Institute of Traditional Chinese Medicine, Guangzhou, China
| |
Collapse
|
8
|
Bighetti-Trevisan RL, Ferraz EP, Silva MBF, Zatta GC, de Almeida MB, Rosa AL, Beloti MM. Effect of osteoblasts on osteoclast differentiation and activity induced by titanium with nanotopography. Colloids Surf B Biointerfaces 2023; 229:113448. [PMID: 37451224 DOI: 10.1016/j.colsurfb.2023.113448] [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: 05/12/2023] [Revised: 07/03/2023] [Accepted: 07/08/2023] [Indexed: 07/18/2023]
Abstract
Titanium with nanotopography (Ti Nano) favors osteoblast differentiation and attenuates the osteoclast inhibitory effects on osteoblasts. Because the interactions between nanotopography and osteoclasts are underexplored, the aims of this study were to evaluate the effects of Ti Nano on osteoclast differentiation and activity, and the influence of osteoblasts on osteoclast-Ti Nano interaction. The discs were conditioned with a mixture of 10 N H2SO4 and 30% aqueous H2O2 to create Ti Nano and non-conditioned Ti discs were used as control (Ti Control). Osteoclasts were cultured on Ti Control and Ti Nano in the presence of osteoblasts in an indirect co-culture system. Also, osteoclasts were cultured on polystyrene and calcium phosphate plates in conditioned media by osteoblasts grown on Ti Control and Ti Nano. While Ti Control exhibited an irregular and smooth surface, Ti Nano presented nanopores distributed throughout the whole surface. Additionally, anisotropy was higher on Ti Nano than Ti Control. Nanotopography favored the gene expression of osteoclast markers but inhibited osteoclast differentiation and activity, and the presence of osteoblasts enhanced the effects of Ti Nano on osteoclasts. Such findings were mimicked by conditioned medium of osteoblasts cultured on Ti Nano, which reduced the osteoclast differentiation and activity. In conclusion, our results indicated that nanotopography regulates osteoblast-osteoclast crosstalk and further investigations should focus the impact of these bone cell interactions on Ti osseointegration.
Collapse
Affiliation(s)
| | - Emanuela Prado Ferraz
- Bone Research Lab, School of Dentistry of Ribeirão Preto, University of São Paulo, Ribeirão Preto 14040-904, SP, Brazil
| | | | - Guilherme Crepi Zatta
- Bone Research Lab, School of Dentistry of Ribeirão Preto, University of São Paulo, Ribeirão Preto 14040-904, SP, Brazil
| | - Marcelo Barros de Almeida
- School of Electrical Engineering, Federal University of Uberlândia, Uberlândia, 38408-100 MG, Brazil
| | - Adalberto Luiz Rosa
- Bone Research Lab, School of Dentistry of Ribeirão Preto, University of São Paulo, Ribeirão Preto 14040-904, SP, Brazil
| | - Marcio Mateus Beloti
- Bone Research Lab, School of Dentistry of Ribeirão Preto, University of São Paulo, Ribeirão Preto 14040-904, SP, Brazil.
| |
Collapse
|
9
|
Li S, Deng Q, Si Q, Li J, Zeng H, Chen S, Guo T. TiO 2nanotubes promote osteogenic differentiation of human bone marrow stem cells via epigenetic regulation of RMRP/ DLEU2/EZH2 pathway. Biomed Mater 2023; 18:055027. [PMID: 37437580 DOI: 10.1088/1748-605x/ace6e9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Accepted: 07/12/2023] [Indexed: 07/14/2023]
Abstract
TiO2nanotubes (TNTs) significantly promote osteogenic differentiation and bone regeneration of cells. Nevertheless, the biological processes by which they promote osteogenesis are currently poorly understood. Long non-coding RNAs (lncRNAs) are essential for controlling osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs). Epigenetic chromatin modification is one of the pathways in which lncRNAs regulate osteogenic differentiation. Here, we reported that TNTs could upregulate lncRNARMRP, and inhibition of lncRNARMRPin human BMSCs (hBMSCs) grown on TNTs could decrease runt-related transcription factor 2 (RUNX2), alkaline phosphatase, osteopontin, and osteocalcin (OCN) expression. Furthermore, we discovered that inhibiting lncRNARMRPelevated the expression of lncRNADLEU2, and lncRNADLEU2knockdown promoted osteogenic differentiation in hBMSCs. RNA immunoprecipitation experiments showed that lncRNADLEU2could interact with EZH2 to induce H3K27 methylation in the promoter regions of RUNX2 and OCN, suppressing gene expression epigenetically. According to these results, lncRNARMRPis upregulated by TNTs to promote osteogenic differentiation throughDLEU2/EZH2-mediated epigenetic modifications.
Collapse
Affiliation(s)
- Shuangqin Li
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu 610031, Sichuan, People's Republic of China
| | - Qing Deng
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu 610031, Sichuan, People's Republic of China
| | - Qiqi Si
- School of Life and Engineering, Southwest Jiaotong University, Chengdu, Sichuan 610031, People's Republic of China
| | - JinSheng Li
- Key Laboratory of Advanced Technologies of Materials Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, People's Republic of China
| | - Huanghe Zeng
- School of Life and Engineering, Southwest Jiaotong University, Chengdu, Sichuan 610031, People's Republic of China
| | - Song Chen
- Department of Orthopedics of the General Hospital of Western Theater Command, Chengdu, Sichuan 610086, People's Republic of China
| | - Tailin Guo
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu 610031, Sichuan, People's Republic of China
| |
Collapse
|
10
|
Si Y, Liu H, Li M, Jiang X, Yu H, Sun D. An efficient metal-organic framework-based drug delivery platform for synergistic antibacterial activity and osteogenesis. J Colloid Interface Sci 2023; 640:521-539. [PMID: 36878070 DOI: 10.1016/j.jcis.2023.02.149] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 02/18/2023] [Accepted: 02/27/2023] [Indexed: 03/06/2023]
Abstract
Bone implants for clinical application should be endowed with antibacterial activity, biocompatibility, and even osteogenesis-promoting properties. In this work, metal-organic framework (MOF) based drug delivery platform was used to modify titanium implants for improved clinical applicability. Methyl Vanillate@Zeolitic Imidazolate Framework-8 (MV@ZIF-8) was immobilized on the polydopamine (PDA) modified titanium. The sustainable release of the Zn2+ and MV causes substantial oxidative damage to Escherichia coli (E. coli) and Staphylococcus aureus (S. aureus). The increased reactive oxygen species (ROS) significantly up-regulates the expression of oxidative stress and DNA damage response genes. Meanwhile, the structural disruption of lipid membranes caused by the ROS, the damage caused by Zinc active sites and the damage accelerated by the MV are both involved in inhibiting bacterial proliferation. The up-regulated expression of the osteogenic-related genes and proteins indicated that the MV@ZIF-8 could effectively promote the osteogenic differentiation of the human bone mesenchymal stem cells (hBMSCs). RNA sequencing and Western blotting analysis revealed that the MV@ZIF-8 coating activates the canonical Wnt/β-catenin signaling pathway through the regulation of tumor necrosis factor (TNF) pathway, thereby promoting the osteogenic differentiation of the hBMSCs. This work demonstrates a promising application of the MOF-based drug delivery platform in bone tissue engineering.
Collapse
Affiliation(s)
- Yunhui Si
- School of Materials, Sun Yat-sen University, Shenzhen, 518107, PR China
| | - Huanyao Liu
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou 510080, PR China
| | - Mengsha Li
- School of Materials Science and Engineering, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Sun Yat-sen University, Guangzhou, 510006, PR China
| | - Xuzhou Jiang
- School of Materials Science and Engineering, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Sun Yat-sen University, Guangzhou, 510006, PR China; Nanotechnology Research Center, Sun Yat-sen University, Guangzhou 510275, PR China.
| | - Hongying Yu
- School of Materials, Sun Yat-sen University, Shenzhen, 518107, PR China.
| | - Dongbai Sun
- School of Materials Science and Engineering, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Sun Yat-sen University, Guangzhou, 510006, PR China.
| |
Collapse
|
11
|
Fernandes CJDC, da Silva RAF, Wood PF, Ferreira MR, de Almeida GS, de Moraes JF, Bezerra FJ, Zambuzzi WF. Titanium-Enriched Medium Promotes Environment-Induced Epigenetic Machinery Changes in Human Endothelial Cells. J Funct Biomater 2023; 14:jfb14030131. [PMID: 36976055 PMCID: PMC10055987 DOI: 10.3390/jfb14030131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 02/21/2023] [Accepted: 02/22/2023] [Indexed: 03/02/2023] Open
Abstract
It is important to understand whether endothelial cells are epigenetically affected by titanium-enriched media when angiogenesis is required during bone development and it is expected to be recapitulated during osseointegration of biomaterials. To better address this issue, titanium-enriched medium was obtained from incubation of titanium discs for up to 24 h as recommended by ISO 10993-5:2016, and further used to expose human umbilical vein endothelial cells (HUVECs) for up to 72 h, when the samples were properly harvested to allow molecular analysis and epigenetics. In general, our data show an important repertoire of epigenetic players in endothelial cells responding to titanium, reinforcing protein related to the metabolism of acetyl and methyl groups, as follows: Histone deacetylases (HDACs) and NAD-dependent deacetylase sirtuin-1 (Sirt1), DNA methyltransferases (DNMTs) and ten-eleven translocation (TET) methylcytosine dioxygenases, which in conjunction culminate in driving chromatin condensation and the methylation profile of DNA strands, respectively. Taking our data into consideration, HDAC6 emerges as important player of this environment-induced epigenetic mechanism in endothelial cells, while Sirt1 is required in response to stimulation of reactive oxygen species (ROS) production, as its modulation is relevant to vasculature surrounding implanted devices. Collectively, all these findings support the hypothesis that titanium keeps the surrounding microenvironment dynamically active and so affects the performance of endothelial cells by modulating epigenetics. Specifically, this study shows the relevance of HDAC6 as a player in this process, possibly correlated with the cytoskeleton rearrangement of those cells. Furthermore, as those enzymes are druggable, it opens new perspectives to consider the use of small molecules to modulate their activities as a biotechnological tool in order to improve angiogenesis and accelerate bone growth with benefits of a fast recovery time for patients.
Collapse
Affiliation(s)
- Célio Júnior da C. Fernandes
- Laboratory of Bioassays and Cellular Dynamics, Department of Chemical and Biological Sciences, Institute of Biosciences, UNESP—São Paulo State University, Botucatu 18618-970, SP, Brazil
| | - Rodrigo A. Foganholi da Silva
- Department of Dentistry, University of Taubaté, Taubaté 12020-340, SP, Brazil
- Program in Environmental and Experimental Pathology, Paulista University, São Paulo 04026-002, SP, Brazil
| | - Patrícia F. Wood
- Laboratory of Bioassays and Cellular Dynamics, Department of Chemical and Biological Sciences, Institute of Biosciences, UNESP—São Paulo State University, Botucatu 18618-970, SP, Brazil
| | - Marcel Rodrigues Ferreira
- Laboratory of Bioassays and Cellular Dynamics, Department of Chemical and Biological Sciences, Institute of Biosciences, UNESP—São Paulo State University, Botucatu 18618-970, SP, Brazil
| | - Gerson S. de Almeida
- Laboratory of Bioassays and Cellular Dynamics, Department of Chemical and Biological Sciences, Institute of Biosciences, UNESP—São Paulo State University, Botucatu 18618-970, SP, Brazil
| | - Julia Ferreira de Moraes
- Laboratory of Bioassays and Cellular Dynamics, Department of Chemical and Biological Sciences, Institute of Biosciences, UNESP—São Paulo State University, Botucatu 18618-970, SP, Brazil
| | - Fábio J. Bezerra
- Laboratory of Bioassays and Cellular Dynamics, Department of Chemical and Biological Sciences, Institute of Biosciences, UNESP—São Paulo State University, Botucatu 18618-970, SP, Brazil
| | - Willian F. Zambuzzi
- Laboratory of Bioassays and Cellular Dynamics, Department of Chemical and Biological Sciences, Institute of Biosciences, UNESP—São Paulo State University, Botucatu 18618-970, SP, Brazil
- Correspondence:
| |
Collapse
|
12
|
Avery D, Morandini L, Sheakley LS, Shah AH, Bui L, Abaricia JO, Olivares-Navarrete R. Canonical Wnt signaling enhances pro-inflammatory response to titanium by macrophages. Biomaterials 2022; 289:121797. [PMID: 36156410 PMCID: PMC10262842 DOI: 10.1016/j.biomaterials.2022.121797] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 08/22/2022] [Accepted: 09/05/2022] [Indexed: 11/19/2022]
Abstract
Biomaterial characteristics like surface roughness and wettability can determine the phenotype of macrophages following implantation. We have demonstrated that inhibiting Wnt ligand secretion abolishes macrophage polarization in vitro and in vivo; however, the role of canonical Wnt signaling in macrophage activation in response to physical and chemical biomaterial cues is unknown. The aim of this study was to understand whether canonical Wnt signaling affects the response of macrophages to titanium (Ti) surface roughness or wettability in vitro and in vivo. Activating canonical Wnt signaling increased expression of toll-like receptors and interleukin receptors and secreted pro-inflammatory cytokines and reduced anti-inflammatory cytokines on Ti, regardless of surface properties. Inhibiting canonical Wnt signaling reduced pro-inflammatory cytokines on all Ti surfaces and increased anti-inflammatory cytokines on rough or rough-hydrophilic Ti. In vivo, activating canonical Wnt signaling increased total macrophages, pro-inflammatory macrophages, and T cells and decreased anti-inflammatory macrophages on both smooth and rough-hydrophilic implants. Functionally, canonical Wnt activation increases pro-inflammatory macrophage response to cell and cell-extracellular matrix lysates. These results demonstrate that activating canonical Wnt signaling primes macrophages to a pro-inflammatory phenotype that affects their response to Ti implants in vitro and in vivo.
Collapse
Affiliation(s)
- Derek Avery
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, Richmond, VA, United States
| | - Lais Morandini
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, Richmond, VA, United States
| | - Luke S Sheakley
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, Richmond, VA, United States
| | - Arth H Shah
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, Richmond, VA, United States
| | - Loc Bui
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, Richmond, VA, United States
| | - Jefferson O Abaricia
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, Richmond, VA, United States
| | - Rene Olivares-Navarrete
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, Richmond, VA, United States.
| |
Collapse
|
13
|
Scannavino RCP, Riccucci G, Ferraris S, Duarte GLC, de Oliveira PT, Spriano S. Functionalization with Polyphenols of a Nano-Textured Ti Surface through a High-Amino Acid Medium: A Chemical-Physical and Biological Characterization. NANOMATERIALS (BASEL, SWITZERLAND) 2022; 12:2916. [PMID: 36079954 PMCID: PMC9458157 DOI: 10.3390/nano12172916] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 08/20/2022] [Accepted: 08/22/2022] [Indexed: 06/15/2023]
Abstract
The study aimed to identify an effective mechanism of adsorption of polyphenols on a nano-textured Ti surface and to evaluate the osteogenic differentiation on it. The source of polyphenols was a natural extract from red grape pomace. A chemical etching was used to form an oxide layer with a nanoscale texture on Ti; this layer is hydrophilic, but without hydroxyl groups with high acidic-basic chemical reactivity. The samples were characterized by electron and fluorescence microscopies, UV-Vis spectroscopy, contact angle measurements, zeta potential titration curves, and Folin-Ciocâlteu test. The presence of an adsorbed layer of polyphenols on the functionalized surface, maintaining redox ability, was confirmed by several tests. Consistent with the surface features, the adsorption was maximized by dissolving the extract in a high-amino acid medium, with respect to an inorganic solution, exploiting the high affinity of amino acids for polyphenols and for porous titanium surfaces. The osteogenic differentiation was assessed on an osteoblastic cell line by immunofluorescence, cell viability, expression of key osteoblast markers, and extracellular matrix mineralization. The surfaces functionalized with the extract diluted in the range 1 × 10-5-1 mg/mL resulted in having a greater osteogenic activity for the highest concentration, with lower values of cell viability; higher expression of alkaline phosphatase, bone sialoprotein, and collagen; and lower levels of osteopontin. In conclusion, the functionalization of a nano-textured Ti surface with polyphenols can potentially favor the osteogenic activity of osseointegrated implants.
Collapse
Affiliation(s)
- Rafaella C. P. Scannavino
- School of Dentistry of Ribeirão Preto, University of São Paulo, Ribeirão Preto 14040-904, SP, Brazil
| | - Giacomo Riccucci
- Department of Applied Science and Technology, Politecnico di Torino, 10126 Torino, Italy
| | - Sara Ferraris
- Department of Applied Science and Technology, Politecnico di Torino, 10126 Torino, Italy
| | - Gabriel L. C. Duarte
- School of Dentistry of Ribeirão Preto, University of São Paulo, Ribeirão Preto 14040-904, SP, Brazil
| | - Paulo T. de Oliveira
- School of Dentistry of Ribeirão Preto, University of São Paulo, Ribeirão Preto 14040-904, SP, Brazil
| | - Silvia Spriano
- Department of Applied Science and Technology, Politecnico di Torino, 10126 Torino, Italy
| |
Collapse
|
14
|
Ebrahim N, El-Halim HEA, Helal OK, El-Azab NEE, Badr OAM, Hassouna A, Saihati HAA, Aborayah NH, Emam HT, El-Wakeel HS, Aljasir M, El-Sherbiny M, Sarg NAS, Shaker GA, Mostafa O, Sabry D, Fouly MAK, Forsyth NR, Elsherbiny NM, Salim RF. Effect of bone marrow mesenchymal stem cells-derived exosomes on diabetes-induced retinal injury: Implication of Wnt/ b-catenin signaling pathway. Biomed Pharmacother 2022; 154:113554. [PMID: 35987163 DOI: 10.1016/j.biopha.2022.113554] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 08/01/2022] [Accepted: 08/14/2022] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Diabetic retinopathy (DR) is a serious microvascular complication of diabetes mellitus. Mesenchymal stem cells are currently studied as therapeutic strategy for management of DR. Exosomes, considered as a promising cell-free therapy option, display biological functions similar to those of their parent cells. In retinal development, Wnt/b-catenin signaling provides key cues for functional progression. The present study aimed to evaluate the potential efficacy of bone marrow-derived mesenchymal stem cell-derived exosomes (BM-MSCs-Ex) in diabetes-induced retinal injury via modulation of the Wnt/ b-catenin signaling pathway. METHODS Eighty-one rats were allocated into 6 groups (control, DR, DR + DKK1, DR + exosomes, DR + Wnt3a and DR + exosomes+Wnt3a). Evaluation of each group was via histopathological examination, assessment of gene and/or protein expression concerned with oxidative stress (SOD1, SOD2, Nox2, Nox4, iNOS), inflammation (TNF-α, ICAM-1, NF-κB) and angiogenesis (VEGF, VE-cadherin). RESULTS Results demonstrated that exosomes blocked the wnt/b-catenin pathway in diabetic retina concomitant with significant reduction of features of DR as shown by downregulation of retinal oxidants, upregulation of antioxidant enzymes, suppression of retinal inflammatory and angiogenic markers. These results were further confirmed by histopathological results, fundus examination and optical coherence tomography. Additionally, exosomes ameliorative effects abrogated wnt3a-triggered retinal injury in DR. CONCLUSION Collectively, these data demonstrated that exosomes ameliorated diabetes-induced retinal injury via suppressing Wnt/ b-catenin signaling with subsequent reduction of oxidative stress, inflammation and angiogenesis.
Collapse
Affiliation(s)
- Nesrine Ebrahim
- Department of Histology and Cell Biology, Faculty of Medicine, Benha University, Egypt; Stem Cell Unit, Faculty of Medicine, Benha University, Egypt.
| | | | - Omayma Kamel Helal
- Department of Histology and Cell Biology, Faculty of Medicine, Benha University, Egypt
| | | | - Omnia A M Badr
- Department of Genetics and Genetic Engineering, Faculty of Agriculture, Benha University, Egypt.
| | - Amira Hassouna
- School of Interprofessional Health Studies, Faculty of Health and Environmental Sciences, AUT University, Auckland, New Zealand.
| | - Hajir A Al Saihati
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, University of Hafr Albatin, Saudi Arabia.
| | | | - Hanan Tawfeek Emam
- Department of Clinical Pharmacology, Faculty of Medicine, Benha University, Egypt.
| | - Hend S El-Wakeel
- Department of Physiology, Faculty of Medicine, Benha University, Egypt.
| | - Mohammad Aljasir
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah, Saudi Arabia.
| | - Mohamed El-Sherbiny
- Department of Basic Medical Sciences, College of Medicine, AlMaarefa University, P.O. Box 71666, Riyadh, 11597, Saudi Arabia; Department of Anatomy, Mansoura Faculty of Medicine, Mansoura University, Egypt.
| | - Naglaa A S Sarg
- Department of Anatomy, Benha Faculty of Medicine, Benha University, Egypt.
| | - Gehan Ahmed Shaker
- Department of Medical Physiology, Faculty of Medicine, Mansoura University, Mansoura, Egypt.
| | - Ola Mostafa
- Department of Histology and Cell Biology, Faculty of Medicine, Benha University, Egypt.
| | - Dina Sabry
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Cairo University, Egypt; Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Badr University, Cairo 11562, Egypt.
| | | | - Nicholas Robert Forsyth
- Guy Hilton Research Laboratories, School of Pharmacy and Bioengineering, Faculty of Medicine and Health Sciences, Keele University, Newcastle ST5 5BG, UK.
| | - Nehal M Elsherbiny
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Tabuk, Tabuk, Saudi Arabia; Biochemistry department, Faculty of Pharmacy, Mansoura University, 35516, Mansoura, Egypt.
| | - Rabab F Salim
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Benha University, Egypt.
| |
Collapse
|
15
|
TiO2 Nanocoatings with Controllable Crystal Type and Nanoscale Topography on Zirconia Implants to Accelerate Bone Formation. Bioinorg Chem Appl 2022; 2022:8650659. [PMID: 35529315 PMCID: PMC9068347 DOI: 10.1155/2022/8650659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Accepted: 03/04/2022] [Indexed: 11/18/2022] Open
Abstract
In dentistry, zirconia implants have emerged as a promising alternative for replacing missing teeth due to their superior aesthetic performance and chemical stability. To improve the osseointegration of zirconia implants, modifying their surface with hierarchical micro/nanotopography and bioactive chemical composition are two effective ways. In this work, a microscale topography was prepared on a zirconia surface using hydrofluoric acid etching, and then a 50 nm TiO2 nanocoating was deposited via atomic layer deposition (ALD). Subsequently, an annealing treatment was used to transform the TiO2 from amorphous to anatase and simultaneously generate nanoscale topography. Various investigations into the coating surface morphology, topography, wettability, and chemical composition were carried out using scanning electron microscopy, white light interferometry, contact-angle measurement, X-ray diffraction, and X-ray photoelectron spectroscopy. In addition, in vitro cytocompatibility and osteogenic potential performance of the coatings were evaluated by human bone marrow mesenchymal stem cells (hBMSCs), and in vivo osseointegration performance was assessed in a rat femoral condyle model. Moreover, the possible mechanism was also investigated. The deposition of TiO2 film with/without annealing treatment did not alter the microscale roughness of the zirconia surface, whereas the nanotopography changed significantly after annealing. The in vitro studies revealed that the anatase TiO2 coating with regular wavelike nanostructure could promote the adhesion and proliferation of osteoblasts and further improve the osteogenic potential in vitro and osseointegration in vivo. These positive effects may be caused by nanoscale topography via the canonical Wnt/β-catenin pathway. The results suggest that using ALD in combination with annealing treatment to fabricate a nanotopographic TiO2 coating is a promising way to improve the osteogenic properties of zirconia implants.
Collapse
|
16
|
Wang X, Xin H, Ning X, Zhang Y, Liu F, Zhang Z, Jia X, Guo W, Hong Y, Sui W. Strontium-loaded titanium implant with rough surface modulates osseointegration by changing sfrp4 in canonical and noncanonical Wnt signaling pathways. Biomed Mater 2022; 17. [PMID: 35349988 DOI: 10.1088/1748-605x/ac61fb] [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: 10/27/2021] [Accepted: 03/29/2022] [Indexed: 11/12/2022]
Abstract
A rough morphology and strontium can activate the Wnt pathway to regulate bone mesenchymal stem cells (rBMSCs) osteogenic differentiation, but the mechanism remains unclear. We constructed smooth Ti (ST) surfaces, rough Ti (RT) surfaces subjected to hydrofluoric acid etching, strontium-loaded smooth Ti (ST-Sr) surfaces subjected to magnetron sputtering, and rough strontium-loaded Ti (RT-Sr) surfaces. We systematically studied the in vitro osteogenic differentiation of rBMSCs on these four surfaces by alkaline phosphatase measurement, Alizarin Red staining and PCR. We also investigated whether crosstalk of the canonical and noncanonical Wnt signaling pathways regulated by sfrp4, which is an inhibitor of canonical and noncanonical Wnt, is the underlying mechanism via PCR on rBMSCs in different stages of osteogenic differentiation. We confirmed the effect of sfrp4 through an in vivo sfrp4-siRNA test. The in vitro osteogenic differentiation of rBMSCs decreased in the order RT-Sr, RT, ST-Sr, and ST. Regarding the mechanism, rough morphology and strontium both enhanced the canonical Wnt pathway to promote osseointegration. Additionally, rough morphology can inhibit sfrp4 to activate the noncanonical Wnt pathway, and then, the activated noncanonical Wnt pathway can suppress the canonical Wnt pathway at the early stage of osteogenic differentiation. Strontium continuously enhanced sfrp4 to inhibit the canonical Wnt pathway instead of activating the noncanonical Wnt pathway. Interestingly, the effect of rough morphology on sfrp4 changed from inhibition to enhancement, and the enhancing effect of strontium on sfrp4 was gradually attenuated. The results of the in vivo sfrp4-siRNA test showed that osseointegration decreased in the order RT-Sr, RT-Sr-siRNA, and ST. Our results suggest that the lack of sfrp4 could suppress osseointegration, indicating that sfrp4 acts as a crucial regulatory molecule for the canonical and noncanonical Wnt pathways during the response of rBMSCs to rough morphology and strontium.
Collapse
Affiliation(s)
- Xiaoyi Wang
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Clinical Research Center for Oral Diseases, Fourth Military Medical University School of Stomatology, 145 Changle West Road, Xi 'an, Xi'an, Shaanxi, 710032, CHINA
| | - He Xin
- Fourth Military Medical University, 145 Changle West Road, Xi 'an, Xi'an, 710032, CHINA
| | - Xiaona Ning
- Ophthalmology, Tangdu Hospital Fourth Military Medical University, No. 1 xinsi Road,Xi'an, Xi'an, Shaanxi, 710038, CHINA
| | - Yubohan Zhang
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Clinical Research Center for Oral Diseases, Fourth Military Medical University School of Stomatology, 145 Changle West Road, Xi 'an, Xi'an, Shaanxi, 710032, CHINA
| | - Fuwei Liu
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Clinical Research Center for Oral Diseases, Fourth Military Medical University School of Stomatology, 145 Changle West Road, Xi 'an, Xi'an, Shaanxi, 710032, CHINA
| | - Zhouyang Zhang
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Clinical Research Center for Oral Diseases, Fourth Military Medical University School of Stomatology, 145 Changle West Road, Xi 'an, Xi'an, Shaanxi, 710032, CHINA
| | - Xuelian Jia
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Clinical Research Center for Oral Diseases, Fourth Military Medical University School of Stomatology, 145 Changle West Road, Xi 'an, Xi'an, Shaanxi, 710032, CHINA
| | - Weiwei Guo
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Clinical Research Center for Oral Diseases, Fourth Military Medical University School of Stomatology, 145 Changle West Road, Xi 'an, Xi'an, Shaanxi, 710032, CHINA
| | - Yonglong Hong
- Shenzhen Hospital of Southern Medical University, 1333 XinHu Road, Shenzhen, Shenzhen, Guangdong, 518000, CHINA
| | - Wen Sui
- Shenzhen Hospital of Southern Medical University, 1333 XinHu Road, Shenzhen 518100, Guangdong, China., Shenzhen, 518000, CHINA
| |
Collapse
|
17
|
Guo W, Yang XG, Shi YL, Wang H. The effects and mechanism of paeoniflorin in promoting osteogenic differentiation of MC3T3-E1. J Orthop Surg Res 2022; 17:90. [PMID: 35164817 PMCID: PMC8842535 DOI: 10.1186/s13018-022-02965-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 01/25/2022] [Indexed: 01/19/2023] Open
Abstract
Background The incidence of osteoporosis and osteoporotic fractures is increasing every year. Traditional Chinese Medicine (TCM) can shed new light on the treatment of osteoporosis. This study aimed to explore the role and mechanism of paeoniflorin in promoting osteogenic differentiation of an osteoblast precursor cell line (MC3T3-E1). Methods MC3T3-E1 cells were cultured in osteogenic induction medium (OIM) and OIM combined with different concentrations of paeoniflorin. The optimal dose of paeoniflorin was assessed by a cell counting kit-8 (CCK-8) assay. Then, alkaline phosphatase (ALP) and Alizarin Red S (ARS) staining were performed to assess the osteogenic capacity of paeoniflorin. The transcription of osteogenic genes and the expression of osteogenic proteins were assessed by RT-PCR and Western blotting, respectively. The transcription of Wnt/β-catenin signaling pathway genes and proteins was assessed by RT-PCR and Western blotting, respectively. Finally, Dickkopf-1 (DKK-1), a Wnt/β-catenin signaling pathway inhibitor, was used to identify whether the Wnt/β-catenin signaling pathway was involved in the osteogenic differentiation of paeoniflorin. Osteoclastogenesis in RAW264.7 cells was identified by tartrate-resistant acid phosphatase (TRAP) staining. Results At concentrations ranging from 0.1 to 100 μM, paeoniflorin was not cytotoxic to MC3T3-E1 cells. Paeoniflorin significantly increased the osteogenic differentiation of MC3T3-E1 cells in a dose-dependent manner. Moreover, paeoniflorin significantly increased osteogenic differentiation gene and protein expression. Through bioinformatic analysis, paeoniflorin-affected genes were found to be involved in different signaling pathways, such as the Wnt/β-catenin signaling pathway. Paeoniflorin enhanced β-catenin and CyclinD1 expression compared with that of the control groups. DKK-1 partially reversed the promoting effects of paeoniflorin in promoting osteogenic differentiation of MC3T3-E1 cells. Moreover, paeoniflorin inhibited the osteoclastogenesis of RAW264.7 cells. Conclusion Paeoniflorin promotes osteogenic differentiation in MC3T3-E1 cells by regulating the Wnt/β-catenin pathway. Paeoniflorin is a potential therapeutic agent for the treatment of osteoporosis.
Collapse
|
18
|
Xiao M, Qian H, Lv J, Wang P. Advances in the Study of the Mechanisms of Physiological Root Resorption in Deciduous Teeth. Front Pediatr 2022; 10:850826. [PMID: 35433548 PMCID: PMC9005890 DOI: 10.3389/fped.2022.850826] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Accepted: 02/23/2022] [Indexed: 11/15/2022] Open
Abstract
Physiological root resorption of deciduous teeth is a complex physiological process that is essential for the normal replacement of deciduous teeth and permanent teeth in clinical practice, but its importance is often overlooked due to the presence of permanent teeth. This physiological process includes not only the resorption of hard tissues of deciduous teeth, such as dentin and cementum, but also the elimination of soft tissues, such as pulp and periodontal ligament (PDL). However, the mechanisms of physiological root resorption are not yet clear. In this article, the advances of research on the mechanisms related to physiological root resorption will be reviewed in two main aspects: hard tissues and soft tissues of deciduous teeth, specifically in relation to the effects of inflammatory microenvironment and mechanical stress on the resorption of hard tissues, the repair of hard tissues, and the elimination and the histological events of soft tissues.
Collapse
Affiliation(s)
- Manxue Xiao
- Department of Pediatric Dentistry, Stomatological Hospital, Southern Medical University, Guangzhou, China
| | - Hong Qian
- Department of Pediatric Dentistry, Stomatological Hospital, Southern Medical University, Guangzhou, China
| | - Jingwen Lv
- Department of Pediatric Dentistry, Stomatological Hospital, Southern Medical University, Guangzhou, China
| | - Peixuan Wang
- Department of Pediatric Dentistry, Stomatological Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
19
|
Williams DF. Biocompatibility pathways and mechanisms for bioactive materials: The bioactivity zone. Bioact Mater 2021; 10:306-322. [PMID: 34901548 PMCID: PMC8636667 DOI: 10.1016/j.bioactmat.2021.08.014] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Accepted: 08/07/2021] [Indexed: 12/14/2022] Open
Abstract
This essay analyzes the scientific evidence that forms the basis of bioactive materials, covering the fundamental understanding of bioactivity phenomena and correlation with the mechanisms of biocompatibility of biomaterials. This is a detailed assessment of performance in areas such as bone-induction, cell adhesion, immunomodulation, thrombogenicity and antimicrobial behavior. Bioactivity is the modulation of biological activity by characteristics of the interfacial region that incorporates the material surface and the immediate local host tissue. Although the term ‘bioactive material’ is widely used and has a well understood general meaning, it would be useful now to concentrate on this interfacial region, considered as ‘the bioactivity zone’. Bioactivity phenomena are either due to topographical/micromechanical characteristics, or to biologically active species that are presented in the bioactivity zone. Examples of topographical/micromechanical effects are the modulation of the osteoblast – osteoclast balance, nanotopographical regulation of cell adhesion, and bactericidal nanostructures. Regulation of bioactivity by biologically active species include their influence, especially of metal ions, on signaling pathways in bone formation, the role of cell adhesion molecules and bioactive peptides in cell attachment, macrophage polarization by immunoregulatory molecules and antimicrobial peptides. While much experimental data exists to demonstrate the potential of such phenomena, there are considerable barriers to their effective clinical translation. This essay shows that there is solid scientific evidence of the existence of bioactivity mechanisms that are associated with some types of biomaterials, especially when the material is modified in a manner designed to specifically induce that activity.
Collapse
Affiliation(s)
- David F Williams
- Wake Forest Institute of Regenerative Medicine, 391 Technology Way. Winston-Salem, North Carolina, 27101, USA
| |
Collapse
|
20
|
Wnt/ β-Catenin Pathway Balances Scaffold Degradation and Bone Formation in Tissue-Engineered Laminae. Stem Cells Int 2021; 2021:8359582. [PMID: 34552633 PMCID: PMC8452400 DOI: 10.1155/2021/8359582] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Accepted: 08/23/2021] [Indexed: 01/02/2023] Open
Abstract
Tissue engineering provides a promising way for the regeneration of artificial vertebral laminae. Previous studies have confirmed the feasibility of reconstructing vertebral laminae via hydroxyapatite-collagen I scaffolds and mesenchymal stromal cells. However, there were no studies exploring the degradation of hydroxyapatite-collagen I scaffolds and the function of Wnt/β-catenin pathway in the process. In this study, tissue-engineered laminae (TEL) were constructed by nanohydroxyapatite/collagen I scaffolds and umbilical cord Wharton's Jelly mesenchymal stromal cells (WJ-MSCs). Cell attachment was observed by scanning electron microscopy, and cell viability was confirmed by Live/Dead staining. The rat models were randomly divided into control and β-catenin inhibition groups. Vertebral lamina defect rat models were made on the fifth lumbar vertebrate, and TEL was implanted into the defect site. After 14 weeks, the newborn laminae were harvested for microcomputed tomography, histology, or transcriptional profile analysis. We found that, for the control group, the newborn lamina formation matched with the scaffold degradation and complete newborn laminae formed at the 14th week; for the β-catenin inhibition group, the scaffold degradation rate overrated the lamina formation and no complete artificial laminae were formed at the 14th week. In addition, the osteoclastic genes, such as Cathepsin K or RANKL, in the control groups were significantly lower than the β-catenin inhibition group, and the antiosteoclastic gene, OPG, in the control group was significantly higher than the β-catenin inhibition group. In conclusion, inhibition of Wnt/β-catenin pathway led to speedy scaffold degradation and deferred artificial lamina formation. Wnt/β-catenin pathway played a critical role in maintaining the balance between scaffold degradation and bone formation in the process of vertebral lamina reconstruction.
Collapse
|
21
|
Wang J, Xia Y, Li J, Wang W. miR-129-5p in exosomes inhibits diabetes-associated osteogenesis in the jaw via targeting FZD4. Biochem Biophys Res Commun 2021; 566:87-93. [PMID: 34119828 DOI: 10.1016/j.bbrc.2021.05.072] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 05/20/2021] [Indexed: 01/13/2023]
Abstract
Diabetes mellitus (DM) influence induces poor osseointegration. The osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs) is a critical factor in successful dental implants. Certain microRNAs play important roles during bone development, and others are deregulated in diabetes. This study investigated the roles of miR-129-5p in the osteoblast differentiation regulation. Exosomes containing miR-129-5p inhibited the osteoblast differentiation and was found in the blood of DM rats. The BMSCs isolated from the jaw of rats were used to detect the miR-129-5p expression. Frizzled (FZD) proteins function as receptors for WNT ligands. The FZD4 was the target of miR-129-5p in dual luciferase assay and Western blot. The miR-129-5p inhibited osteoblast differentiation and decreased the osteoblast markers. The exosomes isolated from the blood of DM rats showed more miR-129-5p level. Results suggested that the exosomes containing miR-129-5p maybe regulators of BMSCs in jaw. The collected exosomes containing miR-129-5p showed the inhibition effect in osteoblast differentiation and decreased the expression osteoblastic markers by targeting FZD4/β-catenin signaling pathway. Therefore, the exosomes containing miR-129-5p in DM rats inhibits osteoblast differentiation by targeting FZD4/β-catenin pathway.
Collapse
Affiliation(s)
- Jinhui Wang
- Department of Clinical Laboratory, Xiamen Children's Hospital, Xiamen, China
| | - Yanyun Xia
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jianhao Li
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Wei Wang
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China.
| |
Collapse
|
22
|
He Y, Chen G, Li Y, Li Y, Yi C, Zhang X, Li H, Zeng B, Wang C, Xie W, Zhao W, Yu D. Effect of magnetic graphene oxide on cellular behaviors and osteogenesis under a moderate static magnetic field. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2021; 37:102435. [PMID: 34186257 DOI: 10.1016/j.nano.2021.102435] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 05/13/2021] [Accepted: 06/01/2021] [Indexed: 12/23/2022]
Abstract
The biological behaviors of magnetic graphene oxide (MGO) in a static magnetic field (SMF) are unknown. The current study is to investigate the cellular behaviors, osteogenesis and the mechanism in BMSCs treated with MGO combined with an SMF. Results showed that the synthetic MGO particles were bio-compatible and could significantly improve the osteogenesis of BMSCs under SMFs, as verified by elevated alkaline phosphatase activity, mineralized nodule formation, and expressions of mRNA and protein levels. Under SMF at the same intensity, the addition of graphene oxide to Fe3O4 could increase the osteogenic ability of BMSCs. The Wnt/β-catenin pathway was indicated to be related to the MGO-driven osteogenic behavior of the BMSCs under SMF. Taken together, our findings suggested that MGO under an SMF could promote osteogenesis in BMSCs through the Wnt/β-catenin pathway and hence should attract more attention for practical applications in bone tissue regeneration.
Collapse
Affiliation(s)
- Yi He
- Hospital of Stomatology, Guanghua School of Stomatology, Institute of Stomatological Research, Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Guanhui Chen
- Department of Stomatology, the Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Ye Li
- Hospital of Stomatology, Guanghua School of Stomatology, Institute of Stomatological Research, Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Yiming Li
- Hospital of Stomatology, Guanghua School of Stomatology, Institute of Stomatological Research, Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Chen Yi
- Hospital of Stomatology, Guanghua School of Stomatology, Institute of Stomatological Research, Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Xiliu Zhang
- Hospital of Stomatology, Guanghua School of Stomatology, Institute of Stomatological Research, Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Hongyu Li
- Hospital of Stomatology, Guanghua School of Stomatology, Institute of Stomatological Research, Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Binghui Zeng
- Hospital of Stomatology, Guanghua School of Stomatology, Institute of Stomatological Research, Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Chao Wang
- Hospital of Stomatology, Guanghua School of Stomatology, Institute of Stomatological Research, Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Weihong Xie
- Hospital of Stomatology, Guanghua School of Stomatology, Institute of Stomatological Research, Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Wei Zhao
- Hospital of Stomatology, Guanghua School of Stomatology, Institute of Stomatological Research, Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China.
| | - Dongsheng Yu
- Hospital of Stomatology, Guanghua School of Stomatology, Institute of Stomatological Research, Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
23
|
Li N, Zhang T, He M, Mu Y. MeCP2 attenuates cardiomyocyte hypoxia/reperfusion-induced injury via regulation of the SFRP4/Wnt/β-catenin axis. Biomarkers 2021; 26:363-370. [PMID: 33726573 DOI: 10.1080/1354750x.2021.1903999] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Objective: Methylated CpG binding protein 2 (MeCP2) is closely associated with heart failure, but its role in I/R injury remains unclear. The purpose of this study was to explore the role and underling mechanism of MeCP2 in myocardial I/R injury.Methods: Hypoxia/reperfusion (H/R)-induced H9c2 cardiomyocytes was used to establish an in vitro I/R injury model. Oxidative stress was assessed by measuring reactive oxygen species (ROS) generation, malondialdehyde (MDA) content and superoxide dismutase (SOD) activity. Cell viability and cell cycle arrest were evaluated by the Cell Counting Kit-8 assay and cell cycle assay, respectively. Apoptosis was determined using flow cytometry analysis.Results: The expression of MeCP2 in H9c2 cells was decreased after H/R treatment. The overexpression of MeCP2 inhibited H/R-induced oxidative stress, cell cycle arrest and apoptosis of H9c2 cells. Moreover, MeCP2 inhibited the activation of secreted frizzled related protein 4 (SFRP4)/Wnt/β-catenin axis, and SFRP4 relieved the effect of MeCP2 on oxidative stress, cell cycle arrest and apoptosis in H/R-induced H9c2 cells.Conclusions: MeCP2 attenuated H/R-induced injury in H9c2 cardiomyocytes by modulating the SFRP4/Wnt/β-catenin axis, which suggested that MeCP2 might serve as a therapeutic target of patients with AMI after reperfusion.
Collapse
Affiliation(s)
- Nan Li
- Department of Cardiology, Xi'an Central Hospital, Xi'an, China
| | - Tao Zhang
- Department of Cardiology, Xi'an Central Hospital, Xi'an, China
| | - Mengying He
- Department of Center sterile supply, Xi'an Hospital of Traditional Chinese Medicine, Shaanxi, China
| | - Yudong Mu
- Department of Clinical Laboratory, Shaanxi Provincial Tumor Hospital, Affiliated Hospital of Medical College of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
24
|
Zhang W, Yang Y, Cui B. New perspectives on the roles of nanoscale surface topography in modulating intracellular signaling. CURRENT OPINION IN SOLID STATE & MATERIALS SCIENCE 2021; 25:100873. [PMID: 33364912 PMCID: PMC7751896 DOI: 10.1016/j.cossms.2020.100873] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
The physical properties of biomaterials, such as elasticity, stiffness, and surface nanotopography, are mechanical cues that regulate a broad spectrum of cell behaviors, including migration, differentiation, proliferation, and reprogramming. Among them, nanoscale surface topography, i.e. nanotopography, defines the nanoscale shape and spatial arrangement of surface elements, which directly interact with the cell membranes and stimulate changes in the cell signaling pathways. In biological systems, the effects of nanotopography are often entangled with those of other mechanical and biochemical factors. Precise engineering of 2D nanopatterns and 3D nanostructures with well-defined features has provided a powerful means to study the cellular responses to specific topographic features. In this Review, we discuss efforts in the last three years to understand how nanotopography affects membrane receptor activation, curvature-induced cell signaling, and stem cell differentiation.
Collapse
Affiliation(s)
| | | | - Bianxiao Cui
- Department of Chemistry, Stanford University, ChEM-H/Wu Tsai Neuroscience Research Complex, S285, 290 Jane Stanford Way, Stanford, CA, 94305, United States
| |
Collapse
|
25
|
Wang L, Huang X, Dai T, Xie J, Lv QX, Hou Y, Kong L, Song Y, Liu F. The role of mitochondrial dynamics in the TiO 2 nanotube-accelerated osteogenic differentiation of MC3T3-E1 cells. Biochem Biophys Res Commun 2020; 535:33-38. [PMID: 33340763 DOI: 10.1016/j.bbrc.2020.12.025] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 12/09/2020] [Indexed: 11/18/2022]
Abstract
Nano titanium implants induce osteogenesis, but how osteoblasts respond to this physical stimulation remains unclear. In this study, we tried to reveal the role of the mitochondrial fission-fusion of osteoblasts in response to a nano titanium surface during the process of osteogenesis, which is important for the design of the surface structure of titanium implants. A TiO2 nanotube array (nano titanium, NT) was fabricated by anodization, and a smooth surface (smooth titanium, ST) was used as a control. We investigated changes in the mitochondrial fission-fusion (MFF) dynamics in MC3T3-E1 cells on the NT surface with those on the ST surface by performing transmission electron microscopy (TEM), confocal laser scanning microscope (CLSM) and real-time PCR. At the same time, we also detected changes in the MFF and osteogenic differentiation of MC3T3-E1 cells after DRP1 downregulation with RNA interference. Cells on the NT surface exhibited more mitochondrial fusion than those on the ST surface, and DRP1 was the key regulatory molecule. Interestingly, DRP1 increased for only a short time at the early stage on the NT surface, and when DRP1 was inhibited by siRNA at the early stage, the osteogenic differentiation of MC3T3-E1 cells significantly decreased. In conclusion, DRP1-regulated mitochondrial dynamics played a key role in the nanotopography-accelerated osteogenic differentiation of MC3T3-E1 cells.
Collapse
Affiliation(s)
- Le Wang
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, China
| | - Xin Huang
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, China; School of Stomatology, Qingdao University, Qingdao, 266003, China
| | - Taiqiang Dai
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, China
| | - Jirong Xie
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, China
| | - Qian-Xin Lv
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, China
| | - Yan Hou
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, China
| | - Liang Kong
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, China
| | - Yu Song
- Qingdao Stomatological Hospital, Qingdao University, Qingdao, 266001, China.
| | - Fuwei Liu
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, China.
| |
Collapse
|
26
|
Capellato P, Camargo SEA, Sachs D. Biological Response to Nanosurface Modification on Metallic Biomaterials. Curr Osteoporos Rep 2020; 18:790-795. [PMID: 33085001 DOI: 10.1007/s11914-020-00635-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/09/2020] [Indexed: 12/20/2022]
Abstract
PURPOSE OF REVIEW New biomaterials for biomedical applications have been developed over the past few years. This work summarizes the current cell lines investigations regarding nanosurface modifications to improve biocompatibility and osseointegration. RECENT FINDINGS Material surfaces presenting biomimetic morphology that provides nanoscale architectures have been shown to alter cell/biomaterial interactions. Topographical and biofunctional surface modifications present a positive effect between material and host response. Nanoscale surfaces on titanium have the potential to provide a successful interface for implantable biomedical devices. Future studies need to directly evaluate how the titanium nanoscale materials will perform in in vivo experiments. Biocompatibility should be determined to identify titanium nanoscale as an excellent option for implant procedures.
Collapse
Affiliation(s)
- Patricia Capellato
- Institute of Physics and Chemistry, Unifei- Federal University of Itajubá, Av. BPS, 1303, Itajubá, MG, 37500 903, Brazil.
| | - Samira Esteves Afonso Camargo
- Restorative Dental Sciences, Division of Prosthodontics, University of Florida, College of Dentistry, Gainesville, FL, USA
| | - Daniela Sachs
- Institute of Physics and Chemistry, Unifei- Federal University of Itajubá, Av. BPS, 1303, Itajubá, MG, 37500 903, Brazil
| |
Collapse
|
27
|
Ye G, Bao F, Zhang X, Song Z, Liao Y, Fei Y, Bunpetch V, Heng BC, Shen W, Liu H, Zhou J, Ouyang H. Nanomaterial-based scaffolds for bone tissue engineering and regeneration. Nanomedicine (Lond) 2020; 15:1995-2017. [PMID: 32812486 DOI: 10.2217/nnm-2020-0112] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The global incidence of bone tissue injuries has been increasing rapidly in recent years, making it imperative to develop suitable bone grafts for facilitating bone tissue regeneration. It has been demonstrated that nanomaterials/nanocomposites scaffolds can more effectively promote new bone tissue formation compared with micromaterials. This may be attributed to their nanoscaled structural and topological features that better mimic the physiological characteristics of natural bone tissue. In this review, we examined the current applications of various nanomaterial/nanocomposite scaffolds and different topological structures for bone tissue engineering, as well as the underlying mechanisms of regeneration. The potential risks and toxicity of nanomaterials will also be critically discussed. Finally, some considerations for the clinical applications of nanomaterials/nanocomposites scaffolds for bone tissue engineering are mentioned.
Collapse
Affiliation(s)
- Guo Ye
- Dr Li Dak Sum & Yip Yio Chin Center for Stem Cells & Regenerative Medicine & Department of Orthopedic Surgery of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, PR China.,Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine & Key Laboratory of Tissue Engineering & Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, PR China
| | - Fangyuan Bao
- Dr Li Dak Sum & Yip Yio Chin Center for Stem Cells & Regenerative Medicine & Department of Orthopedic Surgery of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, PR China.,Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine & Key Laboratory of Tissue Engineering & Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, PR China
| | - Xianzhu Zhang
- Dr Li Dak Sum & Yip Yio Chin Center for Stem Cells & Regenerative Medicine & Department of Orthopedic Surgery of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, PR China.,Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine & Key Laboratory of Tissue Engineering & Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, PR China
| | - Zhe Song
- Dr Li Dak Sum & Yip Yio Chin Center for Stem Cells & Regenerative Medicine & Department of Orthopedic Surgery of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, PR China.,Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine & Key Laboratory of Tissue Engineering & Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, PR China
| | - Youguo Liao
- Dr Li Dak Sum & Yip Yio Chin Center for Stem Cells & Regenerative Medicine & Department of Orthopedic Surgery of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, PR China.,Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine & Key Laboratory of Tissue Engineering & Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, PR China
| | - Yang Fei
- Dr Li Dak Sum & Yip Yio Chin Center for Stem Cells & Regenerative Medicine & Department of Orthopedic Surgery of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, PR China
| | - Varitsara Bunpetch
- Dr Li Dak Sum & Yip Yio Chin Center for Stem Cells & Regenerative Medicine & Department of Orthopedic Surgery of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, PR China.,Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine & Key Laboratory of Tissue Engineering & Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, PR China
| | - Boon Chin Heng
- School of Stomatology, Peking University, Beijing, PR China
| | - Weiliang Shen
- Dr Li Dak Sum & Yip Yio Chin Center for Stem Cells & Regenerative Medicine & Department of Orthopedic Surgery of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, PR China.,Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine & Key Laboratory of Tissue Engineering & Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, PR China.,Department of Sports Medicine, Zhejiang University School of Medicine, Hangzhou, PR China.,China Orthopedic Regenerative Medicine Group (CORMed), Hangzhou, PR China
| | - Hua Liu
- Dr Li Dak Sum & Yip Yio Chin Center for Stem Cells & Regenerative Medicine & Department of Orthopedic Surgery of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, PR China.,Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine & Key Laboratory of Tissue Engineering & Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, PR China.,China Orthopedic Regenerative Medicine Group (CORMed), Hangzhou, PR China
| | - Jing Zhou
- Dr Li Dak Sum & Yip Yio Chin Center for Stem Cells & Regenerative Medicine & Department of Orthopedic Surgery of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, PR China.,Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine & Key Laboratory of Tissue Engineering & Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, PR China.,China Orthopedic Regenerative Medicine Group (CORMed), Hangzhou, PR China
| | - Hongwei Ouyang
- Dr Li Dak Sum & Yip Yio Chin Center for Stem Cells & Regenerative Medicine & Department of Orthopedic Surgery of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, PR China.,Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine & Key Laboratory of Tissue Engineering & Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, PR China.,Department of Sports Medicine, Zhejiang University School of Medicine, Hangzhou, PR China.,China Orthopedic Regenerative Medicine Group (CORMed), Hangzhou, PR China
| |
Collapse
|
28
|
Tovani C, Ferreira CR, Simão AMS, Bolean M, Coppeta L, Rosato N, Bottini M, Ciancaglini P, Ramos AP. Characterization of the in Vitro Osteogenic Response to Submicron TiO 2 Particles of Varying Structure and Crystallinity. ACS OMEGA 2020; 5:16491-16501. [PMID: 32685813 PMCID: PMC7364638 DOI: 10.1021/acsomega.0c00900] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 05/13/2020] [Indexed: 06/11/2023]
Abstract
Titanium oxide (TiO2) nano-/microparticles have been widely used in orthopedic and dental sciences because of their excellent mechanical properties, chemical stability, and ability to promote the osseointegration of implants. However, how the structure and crystallinity of TiO2 particles may affect their osteogenic activity remains elusive. Herein, we evaluated the osteogenic response to submicron amorphous, anatase, and rutile TiO2 particles with controlled size and morphology. First, the ability of TiO2 particles to precipitate apatite was assessed in an acellular medium by using a simulated body fluid (SBF). Three days after the addition to SBF, anatase and rutile TiO2 particles induced the precipitation of aggregates of nanoparticles with a platelike morphology, typical for biomimetic apatite. Conversely, amorphous TiO2 particles induced the precipitation of particles with poor Ca/P atomic ratio only after 14 days of exposure to SBF. Next, the osteogenic response to TiO2 particles was assessed in vitro by incubating MC3T3-E1 preosteoblasts with the particles. The viability and mineralization efficiency of osteoblastic cells were maintained in the presence of all the tested TiO2 particles despite the differences in the induction of apatite precipitation in SBF by TiO2 particles with different structures. Analysis of the particles' surface charge and of the proteins adsorbed onto the particles from the culture media suggested that all the tested TiO2 particles acquired a similar biological identity in the culture media. We posited that this phenomenon attenuated potential differences in osteoblast response to amorphous, anatase, and rutile particles. Our study provides an important insight into the complex relationship between the physicochemical properties and function of TiO2 particles and sheds light on their safe use in medicine.
Collapse
Affiliation(s)
- Camila
B. Tovani
- Faculdade
de Filosofia, Ciências e Letras de Ribeirão Preto—Departamento
de Química, Universidade de SãoPaulo, Ribeirão Preto 14040-901, Brazil
| | - Claudio R. Ferreira
- Faculdade
de Filosofia, Ciências e Letras de Ribeirão Preto—Departamento
de Química, Universidade de SãoPaulo, Ribeirão Preto 14040-901, Brazil
| | - Ana Maria S. Simão
- Faculdade
de Filosofia, Ciências e Letras de Ribeirão Preto—Departamento
de Química, Universidade de SãoPaulo, Ribeirão Preto 14040-901, Brazil
| | - Maytê Bolean
- Faculdade
de Filosofia, Ciências e Letras de Ribeirão Preto—Departamento
de Química, Universidade de SãoPaulo, Ribeirão Preto 14040-901, Brazil
| | - Luca Coppeta
- Department
of Occupational Medicine, University of
Rome Tor Vergata, Rome 00133, Italy
| | - Nicola Rosato
- Department
of Experimental Medicine, University of
Rome Tor Vergata, Rome 00133, Italy
| | - Massimo Bottini
- Department
of Experimental Medicine, University of
Rome Tor Vergata, Rome 00133, Italy
- Sanford
Burnham Prebys Medical Discovery Institute, La Jolla, California 92037, United States
| | - Pietro Ciancaglini
- Faculdade
de Filosofia, Ciências e Letras de Ribeirão Preto—Departamento
de Química, Universidade de SãoPaulo, Ribeirão Preto 14040-901, Brazil
| | - Ana Paula Ramos
- Faculdade
de Filosofia, Ciências e Letras de Ribeirão Preto—Departamento
de Química, Universidade de SãoPaulo, Ribeirão Preto 14040-901, Brazil
| |
Collapse
|
29
|
Abuna RPF, Oliveira FS, Adolpho LF, Fernandes RR, Rosa AL, Beloti MM. Frizzled 6 disruption suppresses osteoblast differentiation induced by nanotopography through the canonical Wnt signaling pathway. J Cell Physiol 2020; 235:8293-8303. [PMID: 32239701 DOI: 10.1002/jcp.29674] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Accepted: 03/08/2020] [Indexed: 02/05/2023]
Abstract
This study aimed to investigate if wingless-related integration site (Wnt) signaling participates in the high osteogenic potential of titanium with nanotopography (Ti-Nano). We showed that among the several components of the Wnt signaling pathway, Frizzled 6 (Fzd6) was the transcript most intensely modulated by nanotopography compared with the untreated Ti surface (Ti-Machined). Then, we investigated whether and how Fzd6 participates in the regulation of osteoblast differentiation caused by nanotopography. The Fzd6 silencing with CRISPR-Cas9 transfection in MC3T3-E1 cells induced a more pronounced inhibition of osteoblast differentiation of cells cultured on nanotopography than those cultured on Ti-Machined. The analysis of the expression of calcium-calmodulin-dependent protein kinase II and β-catenin demonstrated that Fzd6 disruption inhibited the osteoblast differentiation induced by Ti-Nano by preventing the activation of Wnt/β-catenin but not that of Wnt/Ca2+ signaling, which is usually triggered by the receptor Fzd6. These findings elucidate the biological function of Fzd6 as a receptor that triggers Wnt/β-catenin signaling and the cellular mechanisms modulated by nanotopography during osteoblast differentiation.
Collapse
Affiliation(s)
- Rodrigo Paolo Flores Abuna
- Bone Research Lab, School of Dentistry of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Fabiola Singaretti Oliveira
- Bone Research Lab, School of Dentistry of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Leticia Faustino Adolpho
- Bone Research Lab, School of Dentistry of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Roger Rodrigo Fernandes
- Bone Research Lab, School of Dentistry of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Adalberto Luiz Rosa
- Bone Research Lab, School of Dentistry of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Marcio Mateus Beloti
- Bone Research Lab, School of Dentistry of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| |
Collapse
|