1
|
Zhang Y, Chen Y, Shi Y, Hu H, Dai Z, Liu Z, Li X. A phase-transited lysozyme coating doped with strontium on titanium surface for bone repairing via enhanced osteogenesis and immunomodulatory. Front Cell Dev Biol 2025; 12:1506671. [PMID: 39834391 PMCID: PMC11743474 DOI: 10.3389/fcell.2024.1506671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Accepted: 12/04/2024] [Indexed: 01/22/2025] Open
Abstract
Introduction Titanium is currently recognized as an excellent orthopedic implant material, but it often leads to poor osseointegration of the implant, and is prone to aseptic loosening leading to implant failure. Therefore, biofunctionalization of titanium surfaces is needed to enhance their osseointegration and immunomodulation properties to reduce the risk of implant loosening. We concluded that the utilization of PTL-Sr is a direct and effective method for the fabrication of multifunctional implants. Methods In this Study, phase-transited lysozyme (PTL) is deposited onto the surface of titanium (Ti) to construct a functional coating and strontium chloride solution was utilized to produce PTL coatings with Sr2+. The characterization of the strontium-doped PTL coatings (PTL-Sr) was tested by scanning electron microscopy (SEM), energy dispersive X-ray spectroscopy (EDX) and inductively coupled plasma atomic emission spectroscopy (ICP-AES). A series of cell and animal experiments were conducted to investigate the biological functions of PTL-Sr coatings. Results The characterization indicates the successful preparation of PTL-Sr coatings. In vitro cellular experiments have demonstrated that it promotes M2 macrophage polarization and reduces inflammatory mediator production while promoting osteogenic differentiation of bone merrow mesenchymal stem cells (BMSCs). The in vivo subcutaneous implantation model demonstrated its good immunomodulatory and angiogenic properties. Discussion Titanium with PTL-Sr coatings promote biomineralization and immunomodulation, which is suitable for orthopedic applications. Further mechanistic exploration and studies using animal models is necessary to enhance the understanding of the clinical applicability of modified titanium.
Collapse
Affiliation(s)
- Yu Zhang
- Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Yu Chen
- Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Yidan Shi
- The High School Attached to Hunan NormalUniversity, Changsha, China
| | - Hongkun Hu
- Department of Orthopedic Surgery, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Zhongyu Dai
- Department of Orthopedic Surgery, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Zhichen Liu
- Department of Orthopedic Surgery, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Xuanan Li
- Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| |
Collapse
|
2
|
Chen Y, Li Y, Wang X, Mo X, Chen Y, Deng Z, Ye X, Yu J. One-Step Gas Foaming Strategy for Constructing Strontium Nanoparticle Decorated 3D Scaffolds: a New Platform for Repairing Critical Bone Defects. ACS APPLIED MATERIALS & INTERFACES 2024; 16:61664-61678. [PMID: 39474891 DOI: 10.1021/acsami.4c13119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2024]
Abstract
The management of critical-sized bone defects poses significant clinical challenges, particularly in the battlefield and trauma-related injuries. However, bone tissue engineering scaffolds that satisfy high porosity and good angiogenic and osteogenic functions are scarce. In this study, 3D nanofiber scaffolds decorated with strontium nanoparticles (3DS-Sr) were fabricated by combining electrospinning and gas foaming. Sodium borohydride (NaBH4) served a dual role as both a reducing and gas-foaming agent, enabling a one-step process for expansion and modification. In vitro experimental results demonstrated that 3DS-Sr possessed an integrated multilayered porous structure. It promoted angiogenesis by upregulating the expression of hypoxia-inducible factor-1α (HIF-1α) protein and phosphorylation of ERK through the sustained release of Sr2+ and created a favorable microenvironment for osteogenesis by activating the Wnt/β-catenin pathway. In vivo experiments indicated that 3DS-Sr promoted cranial bone regeneration by synergistically promoting the effects of vascularization and osteogenesis. In summary, this study proposed a bioactive bone scaffold in a "one stone, two birds" manner, providing a promising strategy for bone defect repair.
Collapse
Affiliation(s)
- Yujie Chen
- Laboratory of Key Technology and Materials in Minimally Invasive Spine Surgery, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, China
- Center for Spinal Minimally Invasive Research, Shanghai Jiao Tong University, Shanghai 200336, China
- Department of Orthopaedics, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, China
| | - Yucai Li
- Laboratory of Key Technology and Materials in Minimally Invasive Spine Surgery, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, China
- Center for Spinal Minimally Invasive Research, Shanghai Jiao Tong University, Shanghai 200336, China
- Department of Orthopaedics, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, China
| | - Xinyi Wang
- Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Songjiang, Shanghai 201620, China
| | - Xiumei Mo
- Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Songjiang, Shanghai 201620, China
| | - Yicheng Chen
- Laboratory of Key Technology and Materials in Minimally Invasive Spine Surgery, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, China
- Center for Spinal Minimally Invasive Research, Shanghai Jiao Tong University, Shanghai 200336, China
- Department of Orthopaedics, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, China
| | - Zijun Deng
- Laboratory of Key Technology and Materials in Minimally Invasive Spine Surgery, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, China
- Center for Spinal Minimally Invasive Research, Shanghai Jiao Tong University, Shanghai 200336, China
- Department of Orthopaedics, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, China
| | - Xiaojian Ye
- Laboratory of Key Technology and Materials in Minimally Invasive Spine Surgery, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, China
- Center for Spinal Minimally Invasive Research, Shanghai Jiao Tong University, Shanghai 200336, China
- Department of Orthopaedics, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, China
| | - Jiangming Yu
- Laboratory of Key Technology and Materials in Minimally Invasive Spine Surgery, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, China
- Center for Spinal Minimally Invasive Research, Shanghai Jiao Tong University, Shanghai 200336, China
- Department of Orthopaedics, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, China
| |
Collapse
|
3
|
Liu Z, Liu H, Liu S, Li B, Liu Y, Luo E. SIRT1 activation promotes bone repair by enhancing the coupling of type H vessel formation and osteogenesis. Cell Prolif 2024; 57:e13596. [PMID: 38211965 PMCID: PMC11150139 DOI: 10.1111/cpr.13596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 12/10/2023] [Accepted: 12/18/2023] [Indexed: 01/13/2024] Open
Abstract
Bone repair is intricately correlated with vascular regeneration, especially of type H vessels. Sirtuin 1 (SIRT1) expression is closely associated with endothelial function and vascular regeneration; however, the role of SIRT1 in enhancing the coupling of type H vessel formation with osteogenesis to promote bone repair needs to be investigated. A co-culture system combining human umbilical vein endothelial cells and osteoblasts was constructed, and a SIRT1 agonist was used to evaluate the effects of SIRT1 activity. The angiogenic and osteogenic capacities of the co-culture system were examined using short interfering RNA. Mouse models with bone defects in the femur or mandible were established to explore changes in type H vessel formation and bone repair following modulated SIRT1 activity. SIRT1 activation augmented the angiogenic and osteogenic capacities of the co-culture system by activating the PI3K/AKT/FOXO1 signalling pathway and did not significantly regulate osteoblast differentiation. Inhibition of the PI3K/AKT/FOXO1 pathway attenuated SIRT1-mediated effects. The SIRT1 activity in bone defects was positively correlated with the formation of type H vessels and bone repair in vivo, whereas SIRT1 inhibition substantially weakened vascular and bone formation. Thus, SIRT1 is crucial to the coupling of type H vessels with osteogenesis during bone repair.
Collapse
Affiliation(s)
- Zhikai Liu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Hanghang Liu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Shibo Liu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Bolun Li
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Yao Liu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - En Luo
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
4
|
García-Casas I, Valor D, Elayoubi H, Montes A, Pereyra C. Morphological 3D Analysis of PLGA/Chitosan Blend Polymer Scaffolds and Their Impregnation with Olive Pruning Residues via Supercritical CO 2. Polymers (Basel) 2024; 16:1451. [PMID: 38891395 PMCID: PMC11174888 DOI: 10.3390/polym16111451] [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: 04/23/2024] [Revised: 05/16/2024] [Accepted: 05/17/2024] [Indexed: 06/21/2024] Open
Abstract
Natural extracts, such as those from the residues of the Olea europaea industry, offer an opportunity for use due to their richness in antioxidant compounds. These compounds can be incorporated into porous polymeric devices with huge potential for tissue engineering such as bone, cardiovascular, osteogenesis, or neural applications using supercritical CO2. For this purpose, polymeric scaffolds of biodegradable poly(lactic-co-glycolic acid) (PLGA) and chitosan, generated in situ by foaming, were employed for the supercritical impregnation of ethanolic olive leaf extract (OLE). The influence of the presence of chitosan on porosity and interconnectivity in the scaffolds, both with and without impregnated extract, was studied. The scaffolds have been characterized by X-ray computed microtomography, scanning electron microscope, measurements of impregnated load, and antioxidant capacity. The expansion factor decreased as the chitosan content rose, which also occurred when OLE was used. Pore diameters varied, reducing from 0.19 mm in pure PLGA to 0.11 mm in the two experiments with the highest chitosan levels. The connectivity was analyzed, showing that in most instances, adding chitosan doubled the average number of connections, increasing it by a factor of 2.5. An experiment was also conducted to investigate the influence of key factors in the impregnation of the extract, such as pressure (10-30 MPa), temperature (308-328 K), and polymer ratio (1:1-9:1 PLGA/chitosan). Increased pressure facilitated increased OLE loading. The scaffolds were evaluated for antioxidant activity and demonstrated substantial oxidation inhibition (up to 82.5% under optimal conditions) and remarkable potential to combat oxidative stress-induced pathologies.
Collapse
Affiliation(s)
| | - Diego Valor
- Department of Chemical Engineering and Food Technology, Faculty of Sciences, International Excellence, Agrifood Campus (CeiA3), University of Cádiz, 11510 Puerto Real, Spain; (I.G.-C.); (H.E.); (C.P.)
| | | | - Antonio Montes
- Department of Chemical Engineering and Food Technology, Faculty of Sciences, International Excellence, Agrifood Campus (CeiA3), University of Cádiz, 11510 Puerto Real, Spain; (I.G.-C.); (H.E.); (C.P.)
| | | |
Collapse
|
5
|
Cui Y, Hong S, Jiang W, Li X, Zhou X, He X, Liu J, Lin K, Mao L. Engineering mesoporous bioactive glasses for emerging stimuli-responsive drug delivery and theranostic applications. Bioact Mater 2024; 34:436-462. [PMID: 38282967 PMCID: PMC10821497 DOI: 10.1016/j.bioactmat.2024.01.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 12/17/2023] [Accepted: 01/02/2024] [Indexed: 01/30/2024] Open
Abstract
Mesoporous bioactive glasses (MBGs), which belong to the category of modern porous nanomaterials, have garnered significant attention due to their impressive biological activities, appealing physicochemical properties, and desirable morphological features. They hold immense potential for utilization in diverse fields, including adsorption, separation, catalysis, bioengineering, and medicine. Despite possessing interior porous structures, excellent morphological characteristics, and superior biocompatibility, primitive MBGs face challenges related to weak encapsulation efficiency, drug loading, and mechanical strength when applied in biomedical fields. It is important to note that the advantageous attributes of MBGs can be effectively preserved by incorporating supramolecular assemblies, miscellaneous metal species, and their conjugates into the material surfaces or intrinsic mesoporous networks. The innovative advancements in these modified colloidal inorganic nanocarriers inspire researchers to explore novel applications, such as stimuli-responsive drug delivery, with exceptional in-vivo performances. In view of the above, we outline the fabrication process of calcium-silicon-phosphorus based MBGs, followed by discussions on their significant progress in various engineered strategies involving surface functionalization, nanostructures, and network modification. Furthermore, we emphasize the recent advancements in the textural and physicochemical properties of MBGs, along with their theranostic potentials in multiple cancerous and non-cancerous diseases. Lastly, we recapitulate compelling viewpoints, with specific considerations given from bench to bedside.
Collapse
Affiliation(s)
| | | | | | - Xiaojing Li
- Department of Oral & Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, 200011, China
| | - Xingyu Zhou
- Department of Oral & Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, 200011, China
| | - Xiaoya He
- Department of Oral & Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, 200011, China
| | - Jiaqiang Liu
- Department of Oral & Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, 200011, China
| | - Kaili Lin
- Department of Oral & Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, 200011, China
| | - Lixia Mao
- Department of Oral & Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, 200011, China
| |
Collapse
|
6
|
Liu H, Li K, Guo B, Yuan Y, Ruan Z, Long H, Zhu J, Zhu Y, Chen C. Engineering an injectable gellan gum-based hydrogel with osteogenesis and angiogenesis for bone regeneration. Tissue Cell 2024; 86:102279. [PMID: 38007880 DOI: 10.1016/j.tice.2023.102279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 11/10/2023] [Accepted: 11/19/2023] [Indexed: 11/28/2023]
Abstract
Injectable hydrogels are currently a topic of great interest in bone tissue engineering, which could fill irregular bone defects in a short time and avoid traditional major surgery. Herein, we developed an injectable gellan gum (GG)-based hydrogel for bone defect repair by blending nano-hydroxyapatite (nHA) and magnesium sulfate (MgSO4). In order to acquire an injectable GG-based hydrogel with superior osteogenesis, nHA were blended into GG solution with an optimized proportion. For the aim of endowing this hydrogel capable of angiogenesis, MgSO4 was also incorporated. Physicochemical evaluation revealed that GG-based hydrogel containing 5% nHA (w/v) and 2.5 mM MgSO4 (GG/5%nHA/MgSO4) had appropriate sol-gel transition time, showed a porosity-like structure, and could release magnesium ions for at least 14 days. Rheological studies showed that the GG/5%nHA/MgSO4 hydrogel had a stable structure and repeatable self-healing properties. In-vitro results determined that GG/5%nHA/MgSO4 hydrogel presented superior ability on stimulating bone marrow mesenchymal stem cells (BMSCs) to differentiate into osteogenic linage and human umbilical vein endothelial cells (HUVECs) to generate vascularization. In-vivo, GG/5%nHA/MgSO4 hydrogel was evaluated via a rat cranial defect model, as shown by better new bone formation and more neovascularization invasion. Therefore, the study demonstrated that the new injectable hydrogel, is a favorable bioactive GG-based hydrogel, and provides potential strategies for robust therapeutic interventions to improve the repair of bone defect.
Collapse
Affiliation(s)
- Hongbin Liu
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha 410000, Hunan, China
| | - Kaihu Li
- Department of Orthopaedics, The Second Xiangya Hospital of Central South University, Changsha 410000, Hunan, China
| | - Bin Guo
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha 410000, Hunan, China
| | - Yuhao Yuan
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha 410000, Hunan, China
| | - Zhe Ruan
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha 410000, Hunan, China
| | - Haitao Long
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha 410000, Hunan, China
| | - Jianxi Zhu
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha 410000, Hunan, China
| | - Yong Zhu
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha 410000, Hunan, China.
| | - Can Chen
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha 410000, Hunan, China.
| |
Collapse
|
7
|
Lv N, Zhou Z, Hou M, Hong L, Li H, Qian Z, Gao X, Liu M. Research progress of vascularization strategies of tissue-engineered bone. Front Bioeng Biotechnol 2024; 11:1291969. [PMID: 38312513 PMCID: PMC10834685 DOI: 10.3389/fbioe.2023.1291969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Accepted: 12/06/2023] [Indexed: 02/06/2024] Open
Abstract
The bone defect caused by fracture, bone tumor, infection, and other causes is not only a problematic point in clinical treatment but also one of the hot issues in current research. The development of bone tissue engineering provides a new way to repair bone defects. Many animal experimental and rising clinical application studies have shown their excellent application prospects. The construction of rapid vascularization of tissue-engineered bone is the main bottleneck and critical factor in repairing bone defects. The rapid establishment of vascular networks early after biomaterial implantation can provide sufficient nutrients and transport metabolites. If the slow formation of the local vascular network results in a lack of blood supply, the osteogenesis process will be delayed or even unable to form new bone. The researchers modified the scaffold material by changing the physical and chemical properties of the scaffold material, loading the growth factor sustained release system, and combining it with trace elements so that it can promote early angiogenesis in the process of induced bone regeneration, which is beneficial to the whole process of bone regeneration. This article reviews the local vascular microenvironment in the process of bone defect repair and the current methods of improving scaffold materials and promoting vascularization.
Collapse
Affiliation(s)
- Nanning Lv
- Department of Orthopedic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Department of Orthopedic Surgery, The Second People’s Hospital of Lianyungang Affiliated to Kangda College of Nanjing Medical University, Lianyungang, Jiangsu, China
- Department of Orthopedic Surgery, The Affiliated Lianyungang Clinical College of Xuzhou Medical University, Lianyungang, Jiangsu, China
- Department of Orthopedic Surgery, The Affiliated Lianyungang Clinical College of Jiangsu University, Lianyungang, Jiangsu, China
| | - Zhangzhe Zhou
- Department of Orthopedic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Mingzhuang Hou
- Department of Orthopedic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Lihui Hong
- Department of Orthopedic Surgery, The Second People’s Hospital of Lianyungang Affiliated to Kangda College of Nanjing Medical University, Lianyungang, Jiangsu, China
- Department of Orthopedic Surgery, The Affiliated Lianyungang Clinical College of Xuzhou Medical University, Lianyungang, Jiangsu, China
- Department of Orthopedic Surgery, The Affiliated Lianyungang Clinical College of Jiangsu University, Lianyungang, Jiangsu, China
| | - Hongye Li
- Department of Orthopedic Surgery, The Second People’s Hospital of Lianyungang Affiliated to Kangda College of Nanjing Medical University, Lianyungang, Jiangsu, China
- Department of Orthopedic Surgery, The Affiliated Lianyungang Clinical College of Xuzhou Medical University, Lianyungang, Jiangsu, China
- Department of Orthopedic Surgery, The Affiliated Lianyungang Clinical College of Jiangsu University, Lianyungang, Jiangsu, China
| | - Zhonglai Qian
- Department of Orthopedic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Xuzhu Gao
- Department of Orthopedic Surgery, The Second People’s Hospital of Lianyungang Affiliated to Kangda College of Nanjing Medical University, Lianyungang, Jiangsu, China
- Department of Orthopedic Surgery, The Affiliated Lianyungang Clinical College of Xuzhou Medical University, Lianyungang, Jiangsu, China
- Department of Orthopedic Surgery, The Affiliated Lianyungang Clinical College of Jiangsu University, Lianyungang, Jiangsu, China
| | - Mingming Liu
- Department of Orthopedic Surgery, The Second People’s Hospital of Lianyungang Affiliated to Kangda College of Nanjing Medical University, Lianyungang, Jiangsu, China
- Department of Orthopedic Surgery, The Affiliated Lianyungang Clinical College of Xuzhou Medical University, Lianyungang, Jiangsu, China
- Department of Orthopedic Surgery, The Affiliated Lianyungang Clinical College of Jiangsu University, Lianyungang, Jiangsu, China
| |
Collapse
|
8
|
Murugan SS, Dalavi PA, Surya S, Anil S, Gupta S, Shetty R, Venkatesan J. Fabrication and characterizations of simvastatin-containing mesoporous bioactive glass and molybdenum disulfide scaffold for bone tissue engineering. APL Bioeng 2023; 7:046115. [PMID: 38058994 PMCID: PMC10697724 DOI: 10.1063/5.0172002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Accepted: 10/25/2023] [Indexed: 12/08/2023] Open
Abstract
Due to the limitations of the current treatment approaches of allograft and autograft techniques, treating bone disorders is a significant challenge. To address these shortcomings, a novel biomaterial composite is required. This study presents the preparation and fabrication of a novel biomaterial composite scaffold that combines poly (D, L-lactide-co-glycolide) (PLGA), mesoporous bioactive glass (MBG), molybdenum disulfide (MoS2), and simvastatin (Sim) to address the limitations of current bone grafting techniques of autograft and allograft. The fabricated scaffold of PLGA-MBG-MoS2-Sim composites was developed using a low-cost hydraulic press and salt leaching method, and scanning electron microscopy (SEM) analysis confirmed the scaffolds have a pore size between 143 and 240 μm. The protein adsorption for fabricated scaffolds was increased at 24 h. The water adsorption and retention studies showed significant results on the PLGA-MBG-MoS2-Sim composite scaffold. The biodegradation studies of the PLGA-MBG-MoS2-Sim composite scaffold have shown 54% after 28 days. In vitro, bioactivity evaluation utilizing simulated body fluid studies confirmed the development of bone mineral hydroxyapatite on the scaffolds, which was characterized using x-ray diffraction, Fourier transform infrared, and SEM analysis. Furthermore, the PLGA-MBG-MoS2-Sim composite scaffold is biocompatible with C3H10T1/2 cells and expresses more alkaline phosphatase and mineralization activity. Additionally, in vivo research showed that PLGA-MBG-MoS2-Sim stimulates a higher rate of bone regeneration. These findings highlight the fabricated PLGA-MBG-MoS2-Sim composite scaffold presents a promising solution for the limitations of current bone grafting techniques.
Collapse
Affiliation(s)
- Sesha Subramanian Murugan
- Biomaterials Research Laboratory, Yenepoya Research Centre, Yenepoya (Deemed to be University), Deralakatte, Mangalore, Karnataka 575018, India
| | - Pandurang Appana Dalavi
- Biomaterials Research Laboratory, Yenepoya Research Centre, Yenepoya (Deemed to be University), Deralakatte, Mangalore, Karnataka 575018, India
| | - Suprith Surya
- Advancement Surgical Skill Enhancement Division, Yenepoya (Deemed to be University), Deralakatte, Mangalore, Karnataka 575018, India
| | - Sukumaran Anil
- Department of Dentistry, Oral Health Institute, Hamad Medical Corporation, College of Dental Medicine, Qatar University, Doha, Qatar
| | - Sebanti Gupta
- Biomaterials Research Laboratory, Yenepoya Research Centre, Yenepoya (Deemed to be University), Deralakatte, Mangalore, Karnataka 575018, India
| | - Rohan Shetty
- Department of Surgical Oncology, Yenepoya Medical College Hospital, Mangalore, Karnataka, India
| | - Jayachandran Venkatesan
- Biomaterials Research Laboratory, Yenepoya Research Centre, Yenepoya (Deemed to be University), Deralakatte, Mangalore, Karnataka 575018, India
| |
Collapse
|
9
|
Fanovich MA, Di Maio E, Salerno A. Current Trend and New Opportunities for Multifunctional Bio-Scaffold Fabrication via High-Pressure Foaming. J Funct Biomater 2023; 14:480. [PMID: 37754894 PMCID: PMC10531842 DOI: 10.3390/jfb14090480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Revised: 09/03/2023] [Accepted: 09/11/2023] [Indexed: 09/28/2023] Open
Abstract
Biocompatible and biodegradable foams prepared using the high-pressure foaming technique have been widely investigated in recent decades as porous scaffolds for in vitro and in vivo tissue growth. In fact, the foaming process can operate at low temperatures to load bioactive molecules and cells within the pores of the scaffold, while the density and pore architecture, and, hence, properties of the scaffold, can be finely modulated by the proper selection of materials and processing conditions. Most importantly, the high-pressure foaming of polymers is an ideal choice to limit and/or avoid the use of cytotoxic and tissue-toxic compounds during scaffold preparation. The aim of this review is to provide the reader with the state of the art and current trend in the high-pressure foaming of biomedical polymers and composites towards the design and fabrication of multifunctional scaffolds for tissue engineering. This manuscript describes the application of the gas foaming process for bio-scaffold design and fabrication and highlights some of the most interesting results on: (1) the engineering of porous scaffolds featuring biomimetic porosity to guide cell behavior and to mimic the hierarchical architecture of complex tissues, such as bone; (2) the bioactivation of the scaffolds through the incorporation of inorganic fillers and drugs.
Collapse
Affiliation(s)
- María Alejandra Fanovich
- Institute of Materials Science and Technology (INTEMA), National University of Mar del Plata, National Research Council (CONICET), Mar del Plata 7600, Argentina;
| | - Ernesto Di Maio
- Department of Chemical, Materials and Industrial Production Engineering, University of Naples Federico II, 80125 Naples, Italy;
| | - Aurelio Salerno
- Department of Chemical, Materials and Industrial Production Engineering, University of Naples Federico II, 80125 Naples, Italy;
| |
Collapse
|
10
|
Xu Z, Kusumbe AP, Cai H, Wan Q, Chen J. Type H blood vessels in coupling angiogenesis-osteogenesis and its application in bone tissue engineering. J Biomed Mater Res B Appl Biomater 2023; 111:1434-1446. [PMID: 36880538 DOI: 10.1002/jbm.b.35243] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 02/13/2023] [Accepted: 02/23/2023] [Indexed: 03/08/2023]
Abstract
One specific capillary subtype, termed type H vessel, has been found with unique functional characteristics in coupling angiogenesis with osteogenesis. Researchers have fabricated a variety of tissue engineering scaffolds to enhance bone healing and regeneration through the accumulation of type H vessels. However, only a limited number of reviews discussed the tissue engineering strategies for type H vessel regulation. The object of this review is to summary the current utilizes of bone tissue engineering to regulate type H vessels through various signal pathways including Notch, PDGF-BB, Slit3, HIF-1α, and VEGF signaling. Moreover, we give an insightful overview of recent research progress about the morphological, spatial and age-dependent characteristics of type H blood vessels. Their unique role in tying angiogenesis and osteogenesis together via blood flow, cellular microenvironment, immune system and nervous system are also summarized. This review article would provide an insight into the combination of tissue engineering scaffolds with type H vessels and identify future perspectives for vasculized tissue engineering research.
Collapse
Affiliation(s)
- Zhengyi Xu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- West China School of Stomatology, Sichuan University, Chengdu, China
| | - Anjali P Kusumbe
- Medical Research Council (MRC) Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine (WIMM), University of Oxford, Oxford, UK
| | - He Cai
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- West China School of Stomatology, Sichuan University, Chengdu, China
| | - Qianbing Wan
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- West China School of Stomatology, Sichuan University, Chengdu, China
| | - Junyu Chen
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- West China School of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
11
|
He Y, Liang L, Luo C, Zhang ZY, Huang J. Strategies for in situ tissue engineering of vascularized bone regeneration (Review). Biomed Rep 2023; 18:42. [PMID: 37325184 PMCID: PMC10265129 DOI: 10.3892/br.2023.1625] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 04/03/2023] [Indexed: 06/17/2023] Open
Abstract
Numerous physiological processes occur following bone fracture, including inflammatory cell recruitment, vascularization, and callus formation and remodeling. In particular circumstances, such as critical bone defects or osteonecrosis, the regenerative microenvironment is compromised, rendering endogenous stem/progenitor cells incapable of fully manifesting their reparative potential. Consequently, external interventions, such as grafting or augmentation, are frequently necessary. In situ bone tissue engineering (iBTE) employs cell-free scaffolds that possess microenvironmental cues, which, upon implantation, redirect the behavior of endogenous stem/progenitor cells towards a pro-regenerative inflammatory response and reestablish angiogenesis-osteogenesis coupling. This process ultimately results in vascularized bone regeneration (VBR). In this context, a comprehensive review of the current techniques and modalities in VBR-targeted iBTE technology is provided.
Collapse
Affiliation(s)
- Yijun He
- Department of Osteoarthropathy and Sports Medicine, Guangzhou Panyu Central Hospital, Guangzhou, Guangdong 511400, P.R. China
- Translational Research Centre of Regenerative Medicine and 3D Printing of Guangzhou Medical University, Guangdong Province Engineering Research Center for Biomedical Engineering, State Key Laboratory of Respiratory Disease, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510150, P.R. China
| | - Lin Liang
- Department of Osteoarthropathy and Sports Medicine, Guangzhou Panyu Central Hospital, Guangzhou, Guangdong 511400, P.R. China
| | - Cheng Luo
- Department of Osteoarthropathy and Sports Medicine, Guangzhou Panyu Central Hospital, Guangzhou, Guangdong 511400, P.R. China
| | - Zhi-Yong Zhang
- Translational Research Centre of Regenerative Medicine and 3D Printing of Guangzhou Medical University, Guangdong Province Engineering Research Center for Biomedical Engineering, State Key Laboratory of Respiratory Disease, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510150, P.R. China
| | - Jiongfeng Huang
- Department of Osteoarthropathy and Sports Medicine, Guangzhou Panyu Central Hospital, Guangzhou, Guangdong 511400, P.R. China
| |
Collapse
|
12
|
Behara M, Goudy S. FTY720 in immuno-regenerative and wound healing technologies for muscle, epithelial and bone regeneration. Front Physiol 2023; 14:1148932. [PMID: 37250137 PMCID: PMC10213316 DOI: 10.3389/fphys.2023.1148932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 05/02/2023] [Indexed: 05/31/2023] Open
Abstract
In 2010, the FDA approved the administration of FTY720, S1P lipid mediator, as a therapy to treat relapsing forms of multiple sclerosis. FTY720 was found to sequester pro-inflammatory lymphocytes within the lymph node, preventing them from causing injury to the central nervous system due to inflammation. Studies harnessing the anti-inflammatory properties of FTY720 as a pro-regenerative strategy in wound healing of muscle, bone and mucosal injuries are currently being performed. This in-depth review discusses the current regenerative impact of FTY720 due to its anti-inflammatory effect stratified into an assessment of wound regeneration in the muscular, skeletal, and epithelial systems. The regenerative effect of FTY720 in vivo was characterized in three animal models, with differing delivery mechanisms emerging in the last 20 years. In these studies, local delivery of FTY720 was found to increase pro-regenerative immune cell phenotypes (neutrophils, macrophages, monocytes), vascularization, cell proliferation and collagen deposition. Delivery of FTY720 to a localized wound environment demonstrated increased bone, muscle, and mucosal regeneration through changes in gene and cytokine production primarily by controlling the local immune cell phenotypes. These changes in gene and cytokine production reduced the inflammatory component of wound healing and increased the migration of pro-regenerative cells (neutrophils and macrophages) to the wound site. The application of FTY720 delivery using a biomaterial has demonstrated the ability of local delivery of FTY720 to promote local wound healing leveraging an immunomodulatory mechanism.
Collapse
Affiliation(s)
- Monica Behara
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, United States
| | - Steven Goudy
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, United States
- Department of Otolaryngology, Emory University, Atlanta, GA, United States
| |
Collapse
|
13
|
Ilhan M, Kilicarslan M, Alcigir ME, Bagis N, Ekim O, Orhan K. Clindamycin phosphate and bone morphogenetic protein-7 loaded combined nanoparticle-graft and nanoparticle-film formulations for alveolar bone regeneration - An in vitro and in vivo evaluation. Int J Pharm 2023; 636:122826. [PMID: 36918117 DOI: 10.1016/j.ijpharm.2023.122826] [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: 11/29/2022] [Revised: 03/03/2023] [Accepted: 03/08/2023] [Indexed: 03/14/2023]
Abstract
Commonly utilized techniques for healing alveolar bone destruction such as the use of growth factors, suffering from short half-life, application difficulties, and the ability to achieve bioactivity only in the presence of high doses of growth factor. The sustained release of growth factors through a scaffold-based delivery system offers a promising and innovative tool in dentistry. Furthermore, it is suggested to guide the host response by using antimicrobials together with growth factors to prevent recovery and achieve ideal regeneration. Herein, the aim was to prepare and an in vitro - in vivo evaluation of bone morphogenetic protein 7 (BMP-7) and clindamycin phosphate (CDP) loaded polymeric nanoparticles, and their loading into the alginate-chitosan polyelectrolyte complex film or alloplastic graft to accelerate hard tissue regeneration. PLGA nanoparticles containing CDP and BMP-7, separately or together, were prepared using the double emulsion solvent evaporation technique. Through in vitro assays, it was revealed that spherical particles were homogeneously distributed in the combination formulations, and sustained release could be achieved for >12 weeks with all formulations. Also, results from the micro-CT and histopathological analyses indicated that CDP and BMP-7 loaded nanoparticle-film formulations were more effective in treatment than the nanoparticle loaded grafts.
Collapse
Affiliation(s)
- Miray Ilhan
- Ankara University, Faculty of Pharmacy, Department of Pharmaceutical Technology, 06560 Ankara, Türkiye; Duzce University, Faculty of Pharmacy, Department of Pharmaceutical Technology, 81620 Duzce, Türkiye.
| | - Muge Kilicarslan
- Ankara University, Faculty of Pharmacy, Department of Pharmaceutical Technology, 06560 Ankara, Türkiye.
| | - Mehmet Eray Alcigir
- Kirikkale University, Faculty of Veterinary Medicine, Department of Pathology, 71450 Kirikkale, Türkiye.
| | - Nilsun Bagis
- Ankara University, Faculty of Dentistry, Department of Periodontology, 06560 Ankara, Türkiye.
| | - Okan Ekim
- Ankara University, Faculty of Veterinary Medicine, Department of Anatomy, 06110 Ankara, Türkiye.
| | - Kaan Orhan
- Ankara University, Faculty of Dentistry, Department of Dentomaxillofacial Radiology, 06560 Ankara, Türkiye.
| |
Collapse
|
14
|
Montes A, Valor D, Penabad Y, Domínguez M, Pereyra C, de la Ossa EM. Formation of PLGA-PEDOT: PSS Conductive Scaffolds by Supercritical Foaming. MATERIALS (BASEL, SWITZERLAND) 2023; 16:2441. [PMID: 36984321 PMCID: PMC10057315 DOI: 10.3390/ma16062441] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 03/13/2023] [Accepted: 03/16/2023] [Indexed: 06/18/2023]
Abstract
The usage of conjugated materials for the fabrication of foams intended to be used as therapeutic scaffolds is gaining relevance these days, as they hold certain properties that are not exhibited by other polymer types that have been regularly used until the present. Hence, this work aims to design a specific supercritical CO2 foaming process that would allow the production of porous polymeric devices with improved conductive properties, which would better simulate matrix extracellular conditions when used as therapeutic scaffolds (PLGA-PEDOT:PSS) systems. The effects of pressure, temperature, and contact time on the expansion factor, porosity, mechanical properties, and conductivity of the foam have been evaluated. The foams have been characterized by scanning electron and atomic force microscopies, liquid displacement, PBS degradation test, compression, and resistance to conductivity techniques. Values close to 40% porosity were obtained, with a uniform distribution of polymers on the surface and in the interior, expansion factors of up to 10 orders, and a wide range of conductivity values (2.2 × 10-7 to 1.0 × 10-5 S/cm) and mechanical properties (0.8 to 13.6 MPa Young's modulus in compression test). The conductive and porous scaffolds that have been produced by supercritical CO2 in this study show an interesting potential for tissue engineering and for neural or cardiac tissue regeneration purposes due to the fact that electrical conductivity is a crucial factor for proper cell function and tissue development.
Collapse
Affiliation(s)
- Antonio Montes
- Department of Chemical Engineering and Food Technology, Faculty of Sciences, University of Cadiz, International Excellence Agrifood Campus (CeiA3), 11510 Puerto Real, Cadiz, Spain
| | - Diego Valor
- Department of Chemical Engineering and Food Technology, Faculty of Sciences, University of Cadiz, International Excellence Agrifood Campus (CeiA3), 11510 Puerto Real, Cadiz, Spain
| | - Yaiza Penabad
- Department of Chemical Engineering and Food Technology, Faculty of Sciences, University of Cadiz, International Excellence Agrifood Campus (CeiA3), 11510 Puerto Real, Cadiz, Spain
| | - Manuel Domínguez
- Department Condensed Matter Physics and Institute of Electron Microscopy and Materials, Faculty of Sciences, University of Cadiz, International Excellence Agrifood Campus (CeiA3), 11510 Puerto Real, Cadiz, Spain
| | - Clara Pereyra
- Department of Chemical Engineering and Food Technology, Faculty of Sciences, University of Cadiz, International Excellence Agrifood Campus (CeiA3), 11510 Puerto Real, Cadiz, Spain
| | - Enrique Martínez de la Ossa
- Department of Chemical Engineering and Food Technology, Faculty of Sciences, University of Cadiz, International Excellence Agrifood Campus (CeiA3), 11510 Puerto Real, Cadiz, Spain
| |
Collapse
|
15
|
Zhang X, Chen JY, Pei X, Li YH, Feng H, He ZH, Xie WJ, Pei XB, Zhu Z, Wan QB, Wang J. One-Pot Facile Encapsulation of Dimethyloxallyl Glycine by Nanoscale Zeolitic Imidazolate Frameworks-8 for Enhancing Vascularized Bone Regeneration. Adv Healthc Mater 2023; 12:e2202317. [PMID: 36349826 DOI: 10.1002/adhm.202202317] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 10/31/2022] [Indexed: 11/10/2022]
Abstract
In the process of bone tissue regeneration, regulation of osteogenesis-angiogenesis coupling is of great importance. Therefore, dimethyloxallyl glycine (DMOG) is loaded by nanoscale zeolitic imidazolate frameworks-8 (ZIF-8) to obtain a drug-loading system that can promote osteogenesis-angiogenesis coupling. Characterization of the drug-loading nanoparticles (DMOG@ZIF-8) reveals that DMOG is successfully loaded into ZIF-8 by two different methods, and the DMOG@ZIF-8 is prepared using the one-pot method (OD@ZIF-8) achieves higher loading efficiency and longer release time than those prepared using the post-loading method (PD@ZIF-8). In vitro studies found that DMOG@ZIF-8 significantly enhances the migration, tube formation, and angiogenesis-related protein secretion of human umbilical vein endothelial cells as well as the extracellular matrix mineralization, alkaline phosphatase activity, and osteogenesis-related protein secretion of bone marrow mesenchymal stem cells. Moreover, OD@ZIF-8 nanoparticles are more efficient than PD@ZIF-8 nanoparticles in induction of osteogenesis-angiogenesis coupling. Then, in vivo cranial critical defect model shows that the addition of OD@ZIF-8 significantly promotes vascularized bone formation as indicated by the results including microcomputed tomographic, histological and immunofluorescence staining, and so on. Taken together, loading ZIF-8 with DMOG may be a promising solution for critical-sized bone defect reconstruction and the one-pot method is preferred in the preparation of such drug-loading system.
Collapse
Affiliation(s)
- Xin Zhang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chengdu, 610041, China.,Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Jun-Yu Chen
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chengdu, 610041, China.,Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Xiang Pei
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chengdu, 610041, China
| | - Ya-Hong Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chengdu, 610041, China.,Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Hao Feng
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chengdu, 610041, China.,Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Zi-Han He
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chengdu, 610041, China.,Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Wen-Jia Xie
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chengdu, 610041, China.,Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Xi-Bo Pei
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chengdu, 610041, China.,Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Zhou Zhu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chengdu, 610041, China.,Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Qian-Bing Wan
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chengdu, 610041, China.,Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Jian Wang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chengdu, 610041, China.,Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
16
|
Zhou J, Li Y, He J, Liu L, Hu S, Guo M, Liu T, Liu J, Wang J, Guo B, Wang W. ROS Scavenging Graphene-Based Hydrogel Enhances Type H Vessel Formation and Vascularized Bone Regeneration via ZEB1/Notch1 Mediation. Macromol Biosci 2023; 23:e2200502. [PMID: 36637816 DOI: 10.1002/mabi.202200502] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 01/06/2023] [Indexed: 01/14/2023]
Abstract
The regeneration strategy for bone defects is greatly limited by the bone microenvironment, and excessive reactive oxygen species (ROS) seriously hinder the formation of new bone. Reduced graphene oxide (rGO) is expected to meet the requirements because of its ability to scavenge free radicals through electron transfer. Antioxidant hydrogels based on gelatine methacrylate (GM), acrylyl-β-cyclodextrin (Ac-CD), and rGO functionalized with β-cyclodextrin (β-CD) are developed for skull defect regeneration, but the mechanism of how rGO-based hydrogels enhance bone repair remains unclear. In this work, it is confirmed that the GM/Ac-CD/rGO hydrogel has good antioxidant capacity, and promotes osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs) and angiogenesis of human umbilical vein endothelial cells (HUVECs). The rGO-based hydrogel affects ZEB1/Notch1 to promote tube formation. Furthermore, two-photon laser scanning microscopy is used to observe the ROS in a skull defect. The rGO-based hydrogel promotes type H vessel formation in a skull defect. In conclusion, the hydrogel neutralizes ROS in the vicinity of a skull defect and stimulates ZEB1/Notch1 to promote the coupling of osteogenesis and angiogenesis, which may be a possible approach for bone regeneration.
Collapse
Affiliation(s)
- Junpeng Zhou
- Department of Bone and Joint Surgery, the Second Affiliated Hospital of Xi'an Jiaotong University, NO. 157, Xiwu Road, Xi'an, Shaanxi, 710004, P. R. China
| | - Yongwei Li
- Department of Bone and Joint Surgery, the Second Affiliated Hospital of Xi'an Jiaotong University, NO. 157, Xiwu Road, Xi'an, Shaanxi, 710004, P. R. China
| | - Jiahui He
- State Key Laboratory for Mechanical Behavior of Materials, and Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Liying Liu
- Biomedical Experimental Center of Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710116, China
| | - Shugang Hu
- Department of Bone and Joint Surgery, the Second Affiliated Hospital of Xi'an Jiaotong University, NO. 157, Xiwu Road, Xi'an, Shaanxi, 710004, P. R. China
| | - Meng Guo
- State Key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Tun Liu
- Department of Bone and Joint Surgery, the Second Affiliated Hospital of Xi'an Jiaotong University, NO. 157, Xiwu Road, Xi'an, Shaanxi, 710004, P. R. China
| | - Junzheng Liu
- Department of Bone and Joint Surgery, the Second Affiliated Hospital of Xi'an Jiaotong University, NO. 157, Xiwu Road, Xi'an, Shaanxi, 710004, P. R. China
| | - Jiaxin Wang
- State Key Laboratory for Mechanical Behavior of Materials, and Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Baolin Guo
- State Key Laboratory for Mechanical Behavior of Materials, and Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China.,Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Wei Wang
- Department of Bone and Joint Surgery, the Second Affiliated Hospital of Xi'an Jiaotong University, NO. 157, Xiwu Road, Xi'an, Shaanxi, 710004, P. R. China
| |
Collapse
|
17
|
Zhao H, Liu C, Liu Y, Ding Q, Wang T, Li H, Wu H, Ma T. Harnessing electromagnetic fields to assist bone tissue engineering. Stem Cell Res Ther 2023; 14:7. [PMID: 36631880 PMCID: PMC9835389 DOI: 10.1186/s13287-022-03217-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 12/08/2022] [Indexed: 01/13/2023] Open
Abstract
Bone tissue engineering (BTE) emerged as one of the exceptional means for bone defects owing to it providing mechanical supports to guide bone tissue regeneration. Great advances have been made to facilitate the success of BTE in regenerating bone within defects. The use of externally applied fields has been regarded as an alternative strategy for BTE. Electromagnetic fields (EMFs), known as a simple and non-invasive therapy, can remotely provide electric and magnetic stimulation to cells and biomaterials, thus applying EMFs to assist BTE would be a promising strategy for bone regeneration. When combined with BTE, EMFs improve cell adhesion to the material surface by promoting protein adsorption. Additionally, EMFs have positive effects on mesenchymal stem cells and show capabilities of pro-angiogenesis and macrophage polarization manipulation. These advantages of EMFs indicate that it is perfectly suitable for representing the adjuvant treatment of BTE. We also summarize studies concerning combinations of EMFs and diverse biomaterial types. The strategy of combining EMFs and BTE receives encouraging outcomes and holds a promising future for effectively treating bone defects.
Collapse
Affiliation(s)
- Hongqi Zhao
- grid.33199.310000 0004 0368 7223Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 Hubei China
| | - Chaoxu Liu
- grid.33199.310000 0004 0368 7223Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 Hubei China
| | - Yang Liu
- grid.33199.310000 0004 0368 7223Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 Hubei China
| | - Qing Ding
- grid.33199.310000 0004 0368 7223Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 Hubei China
| | - Tianqi Wang
- grid.33199.310000 0004 0368 7223Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 Hubei China
| | - Hao Li
- grid.33199.310000 0004 0368 7223Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 Hubei China
| | - Hua Wu
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China.
| | - Tian Ma
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China.
| |
Collapse
|
18
|
Novel structural designs of 3D-printed osteogenic graft for rapid angiogenesis. Biodes Manuf 2022. [DOI: 10.1007/s42242-022-00212-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
19
|
You J, Zhang Y, Zhou Y. Strontium Functionalized in Biomaterials for Bone Tissue Engineering: A Prominent Role in Osteoimmunomodulation. Front Bioeng Biotechnol 2022; 10:928799. [PMID: 35875505 PMCID: PMC9298737 DOI: 10.3389/fbioe.2022.928799] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 06/13/2022] [Indexed: 12/24/2022] Open
Abstract
With the development of bone tissue engineering bio-scaffold materials by adding metallic ions to improve bone healing have been extensively explored in the past decades. Strontium a non-radioactive element, as an essential osteophilic trace element for the human body, has received widespread attention in the medical field due to its superior biological properties of inhibiting bone resorption and promoting osteogenesis. As the concept of osteoimmunology developed, the design of orthopedic biomaterials has gradually shifted from “immune-friendly” to “immunomodulatory” with the aim of promoting bone healing by modulating the immune microenvironment through implanted biomaterials. The process of bone healing can be regarded as an immune-induced procedure in which immune cells can target the effector cells such as macrophages, neutrophils, osteocytes, and osteoprogenitor cells through paracrine mechanisms, affecting pathological alveolar bone resorption and physiological bone regeneration. As a kind of crucial immune cell, macrophages play a critical role in the early period of wound repair and host defense after biomaterial implantation. Despite Sr-doped biomaterials being increasingly investigated, how extracellular Sr2+ guides the organism toward favorable osteogenesis by modulating macrophages in the bone tissue microenvironment has rarely been studied. This review focuses on recent knowledge that the trace element Sr regulates bone regeneration mechanisms through the regulation of macrophage polarization, which is significant for the future development of Sr-doped bone repair materials. We will also summarize the primary mechanism of Sr2+ in bone, including calcium-sensing receptor (CaSR) and osteogenesis-related signaling pathways.
Collapse
Affiliation(s)
- Jiaqian You
- Department of Oral Implantology, Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, China
| | - Yidi Zhang
- Department of Oral Implantology, Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, China
| | - Yanmin Zhou
- Department of Oral Implantology, Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, China
| |
Collapse
|
20
|
Yi Y, Hu WJ, Zhao CR, Xiong MC, Zhang Q, Wu YP, Zeng H, Zeng N. The protective role of FTY720 in promoting survival of allograft fat in mice. Kaohsiung J Med Sci 2022; 38:889-896. [PMID: 35833419 DOI: 10.1002/kjm2.12570] [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: 10/18/2021] [Revised: 05/19/2022] [Accepted: 05/25/2022] [Indexed: 11/07/2022] Open
Abstract
Fat transplantation is widely used for soft-tissue filling and wound repair. Owing to the biological changes in adipocytes in some metabolic diseases, allograft fat can provide a better source of donor fat than autologous fat. Fingolimod (FTY720) possesses a powerful immunomodulatory function. This study aimed to investigate the protective effect of FTY720 in allogeneic fat transplantation. C57BL/6J mice that received allografts were randomly divided into two groups and treated with saline and FTY720, respectively. Fat graft samples were obtained at 1, 6, and 20 weeks posttransplantation. Graft volumes, graft structure, and immune cells were estimated using histological examination, immunohistochemistry, staining immunofluorescence (IF), and quantitative real-time polymerase chain reaction (qRT-PCR). Inflammatory cytokine mRNA expression in grafts was detected by qRT-PCR. FTY720 treatment significantly enhanced allograft retention, structural integrity, and neovascularization, thereby demonstrating the potential of FTY720 in improving graft survival. Further IF staining showed that FTY720 increased regulatory T cell infiltration and reduced macrophage infiltration to some extent. FTY720 treatment also enhanced the expression of the anti-inflammatory cytokines interleukin (IL)-4 and IL-10 and weakened the expression of the pro-inflammatory cytokines TNF-α and IL-6. Furthermore, FTY720 treatment upregulated the expression of CD31 positive cells. This study demonstrated the potential efficacy of FTY720 in improving the graft survival rate of syngeneic fat allograft models, possibly by suppressing immune rejection and promoting angiogenesis. Therefore, this study offers key insights into the potential application of a drug-assisted strategy to prolong allograft fat survival.
Collapse
Affiliation(s)
- Yi Yi
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Wei-Jie Hu
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Chong-Ru Zhao
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Ming-Chen Xiong
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Qi Zhang
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yi-Ping Wu
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Hong Zeng
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Ning Zeng
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
21
|
Identification of Type-H-like Blood Vessels in a Dynamic and Controlled Model of Osteogenesis in Rabbit Calvarium. MATERIALS 2022; 15:ma15134703. [PMID: 35806828 PMCID: PMC9267487 DOI: 10.3390/ma15134703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Revised: 06/17/2022] [Accepted: 07/01/2022] [Indexed: 02/06/2023]
Abstract
Angiogenesis and bone regeneration are closely interconnected processes. Whereas type-H blood vessels are abundantly found in the osteogenic zones during endochondral long bone development, their presence in flat bones’ development involving intramembranous mechanisms remains unclear. Here, we hypothesized that type-H-like capillaries that highly express CD31 and Endomucin (EMCN), may be present at sites of intramembranous bone development and participate in the control of osteogenesis. A rabbit model of calvarial bone augmentation was used in which bone growth was controlled over time (2–4 weeks) using a particulate bone scaffold. The model allowed the visualization of the entire spectrum of stages throughout bone growth in the same sample, i.e., active ossification, osteogenic activity, and controlled inflammation. Using systematic mRNA hybridization, the formation of capillaries subpopulations (CD31–EMCN staining) over time was studied and correlated with the presence of osteogenic precursors (Osterix staining). Type-H-like capillaries strongly expressing CD31 and EMCN were identified and described. Their presence increased gradually from the regenerative zone up to the osteogenic zone, at 2 and 4 weeks. Type-H-like capillaries may thus represent the initial vascular support encountered in flat bones’ development and which organize osteogenic niches.
Collapse
|
22
|
Bai H, Wang Y, Zhao Y, Chen X, Xiao Y, Bao C. HIF signaling: A new propellant in bone regeneration. BIOMATERIALS ADVANCES 2022; 138:212874. [PMID: 35913258 DOI: 10.1016/j.bioadv.2022.212874] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 05/04/2022] [Accepted: 05/16/2022] [Indexed: 06/15/2023]
Abstract
Bone tissue destruction leads to severe pain, physical flaws, and loss of motility. Bone repair using biocompatible and osteo-inductive scaffolds is regarded as a viable and potential therapeutic approach. However, for large-scale bone regeneration, oxygen and nutrient supply have become limiting factors. Further, a considerable need exists for recruited cell activities and blood vessel growth. Hypoxia-inducible factor (HIF) signaling pathways induced by hypoxia are involved in angiogenesis and osteogenesis. As an important transcription factor, HIF-1 functions by modulating vital genes, such as VEGF, PDK1, and EPO, and is a crucial regulator that influences the final fate of bone regeneration. Collectively, to achieve better osteogenesis results, the in-depth molecular mechanisms that underpin the links between materials, cells, and HIF signaling pathways must be determined. This review aimed to provide an in-depth insight into recent progress in HIF-regulated bone regeneration. Hypoxia and cellular oxygen-sensing mechanisms and their correlations with osteogenesis were determined, and recent studies on hypoxia-inducing and hypoxia-mimicking strategies were briefly described. Finally, the potential applications of HIF signaling in bone regeneration were highlighted. This review provides theoretical support for establishing a novel and viable bone repair strategy in the clinic by harnessing HIF signaling.
Collapse
Affiliation(s)
- Hetian Bai
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Med-X Center for Materials, Sichuan University, No. 14, Section 3, Renmin Nan Road, Chengdu 610041, Sichuan, China
| | - Yue Wang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Med-X Center for Materials, Sichuan University, No. 14, Section 3, Renmin Nan Road, Chengdu 610041, Sichuan, China
| | - Yi Zhao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Med-X Center for Materials, Sichuan University, No. 14, Section 3, Renmin Nan Road, Chengdu 610041, Sichuan, China
| | - Xin Chen
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Med-X Center for Materials, Sichuan University, No. 14, Section 3, Renmin Nan Road, Chengdu 610041, Sichuan, China
| | - Yu Xiao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Med-X Center for Materials, Sichuan University, No. 14, Section 3, Renmin Nan Road, Chengdu 610041, Sichuan, China.
| | - Chongyun Bao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Med-X Center for Materials, Sichuan University, No. 14, Section 3, Renmin Nan Road, Chengdu 610041, Sichuan, China
| |
Collapse
|
23
|
Chen A, Li X, Zhao J, Zhou J, Xie C, Chen H, Wang Q, Wang R, Miao D, Li J, Jin J. Chronic alcohol reduces bone mass through inhibiting proliferation and promoting aging of endothelial cells in type-H vessels. Stem Cells Dev 2022; 31:541-554. [DOI: 10.1089/scd.2021.0337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Ao Chen
- Nanjing Medical University, 12461, Research Centre for Bone and Stem Cells, Department of Human Anatomy; Key Laboratory for Aging & Disease; The State Key Laboratory of Reproductive Medicine, Nanjing, Jiangsu, China
| | - Xiaoting Li
- Nanjing Medical University, 12461, Department of Nutrition and Food Safety, School of Public Health, Nanjing, Jiangsu, China
| | - Jingyu Zhao
- Nanjing Medical University, 12461, Research Centre for Bone and Stem Cells, Department of Human Anatomy; Key Laboratory for Aging & Disease; The State Key Laboratory of Reproductive Medicine, Nanjing, Jiangsu, China
| | - Jiawen Zhou
- Nanjing Medical University, 12461, Research Centre for Bone and Stem Cells, Department of Human Anatomy; Key Laboratory for Aging & Disease; The State Key Laboratory of Reproductive Medicine, Nanjing, Jiangsu, China
| | - Chunfeng Xie
- Nanjing Medical University, 12461, Department of Nutrition and Food Safety, School of Public Health, Nanjing, Jiangsu, China
| | - Haiyun Chen
- Nanjing Medical University, 12461, Anti-aging Research Laboratory, Friendship Plastic Surgery Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Qiuyi Wang
- Nanjing Medical University, 12461, Research Centre for Bone and Stem Cells, Department of Human Anatomy; Key Laboratory for Aging & Disease; The State Key Laboratory of Reproductive Medicine, Nanjing, Jiangsu, China
| | - Rong Wang
- Nanjing Medical University, 12461, Research Centre for Bone and Stem Cells, Department of Human Anatomy; Key Laboratory for Aging & Disease; The State Key Laboratory of Reproductive Medicine, Nanjing, Jiangsu, China
| | - Dengshun Miao
- Nanjing Medical University, Nanjing, Jiangsu, China, 210029, ,
| | - Jie Li
- Xuzhou Medical University, 38044, Department of Orthopaedics, Xuzhou Central Hospital; The Xuzhou Clinical School of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Jianliang Jin
- Nanjing Medical University, 12461, Nanjing, China, 211166
- No.101,Longmian Avenue,Jiangning DistrictChina
| |
Collapse
|
24
|
Gonzalez-Vilchis RA, Piedra-Ramirez A, Patiño-Morales CC, Sanchez-Gomez C, Beltran-Vargas NE. Sources, Characteristics, and Therapeutic Applications of Mesenchymal Cells in Tissue Engineering. Tissue Eng Regen Med 2022; 19:325-361. [PMID: 35092596 PMCID: PMC8971271 DOI: 10.1007/s13770-021-00417-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 11/24/2021] [Accepted: 12/05/2021] [Indexed: 01/31/2023] Open
Abstract
Tissue engineering (TE) is a therapeutic option within regenerative medicine that allows to mimic the original cell environment and functional organization of the cell types necessary for the recovery or regeneration of damaged tissue using cell sources, scaffolds, and bioreactors. Among the cell sources, the utilization of mesenchymal cells (MSCs) has gained great interest because these multipotent cells are capable of differentiating into diverse tissues, in addition to their self-renewal capacity to maintain their cell population, thus representing a therapeutic alternative for those diseases that can only be controlled with palliative treatments. This review aimed to summarize the state of the art of the main sources of MSCs as well as particular characteristics of each subtype and applications of MSCs in TE in seven different areas (neural, osseous, epithelial, cartilage, osteochondral, muscle, and cardiac) with a systemic revision of advances made in the last 10 years. It was observed that bone marrow-derived MSCs are the principal type of MSCs used in TE, and the most commonly employed techniques for MSCs characterization are immunodetection techniques. Moreover, the utilization of natural biomaterials is higher (41.96%) than that of synthetic biomaterials (18.75%) for the construction of the scaffolds in which cells are seeded. Further, this review shows alternatives of MSCs derived from other tissues and diverse strategies that can improve this area of regenerative medicine.
Collapse
Affiliation(s)
- Rosa Angelica Gonzalez-Vilchis
- Molecular Biology Undergraduate Program, Natural Science and Engineering Division, Cuajimalpa Unit, Autonomous Metropolitan University, 05340, CDMX, Mexico
| | - Angelica Piedra-Ramirez
- Molecular Biology Undergraduate Program, Natural Science and Engineering Division, Cuajimalpa Unit, Autonomous Metropolitan University, 05340, CDMX, Mexico
| | - Carlos Cesar Patiño-Morales
- Research Laboratory of Developmental Biology and Experimental Teratogenesis, Children's Hospital of Mexico Federico Gomez, 06720, CDMX, Mexico
| | - Concepcion Sanchez-Gomez
- Research Laboratory of Developmental Biology and Experimental Teratogenesis, Children's Hospital of Mexico Federico Gomez, 06720, CDMX, Mexico
| | - Nohra E Beltran-Vargas
- Department of Processes and Technology, Natural Science and Engineering Division, Cuajimalpa Unit, Autonomous Metropolitan University, Cuajimalpa. Vasco de Quiroga 4871. Cuajimalpa de Morelos, 05348, CDMX, Mexico.
| |
Collapse
|
25
|
Zhang Y, Wang J, Zhou J, Sun J, Jiao Z. Multi‐modal cell structure formation of poly (lactic‐co‐glycolic acid)/superparamagnetic iron oxide nanoparticles composite scaffolds by supercritical
CO
2
varying‐temperature foaming. POLYM ADVAN TECHNOL 2022. [DOI: 10.1002/pat.5650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Affiliation(s)
- Yi Zhang
- School of Energy and Environment Southeast University Nanjing Jiangsu China
- Jiangsu Key Laboratory for Biomaterials and Devices Southeast University Nanjing Jiangsu China
- Joint Research Institute of Southeast University and Monash University Southeast University Suzhou Jiangsu China
| | - Jinjing Wang
- School of Energy and Environment Southeast University Nanjing Jiangsu China
- Jiangsu Key Laboratory for Biomaterials and Devices Southeast University Nanjing Jiangsu China
- Joint Research Institute of Southeast University and Monash University Southeast University Suzhou Jiangsu China
| | - Jiancheng Zhou
- School of Chemistry and Chemical Engineering Southeast University Nanjing Jiangsu China
| | - Jianfei Sun
- Jiangsu Key Laboratory for Biomaterials and Devices Southeast University Nanjing Jiangsu China
- Joint Research Institute of Southeast University and Monash University Southeast University Suzhou Jiangsu China
| | - Zhen Jiao
- Jiangsu Key Laboratory for Biomaterials and Devices Southeast University Nanjing Jiangsu China
- Joint Research Institute of Southeast University and Monash University Southeast University Suzhou Jiangsu China
- School of Chemistry and Chemical Engineering Southeast University Nanjing Jiangsu China
| |
Collapse
|
26
|
Duarte MM, Silva IV, Eisenhut AR, Bionda N, Duarte ARC, Oliveira AL. Contributions of supercritical fluid technology for advancing decellularization and postprocessing of viable biological materials. MATERIALS HORIZONS 2022; 9:864-891. [PMID: 34931632 DOI: 10.1039/d1mh01720a] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
The demand for tissue and organ transplantation worldwide has led to an increased interest in the development of new therapies to restore normal tissue function through transplantation of injured tissue with biomedically engineered matrices. Among these developments is decellularization, a process that focuses on the removal of immunogenic cellular material from a tissue or organ. However, decellularization is a complex and often harsh process that frequently employs techniques that can negatively impact the properties of the materials subjected to it. The need for a more benign alternative has driven research on supercritical carbon dioxide (scCO2) assisted decellularization. scCO2 can achieve its critical point at relatively low temperature and pressure conditions, and for its high transfer rate and permeability. These properties make scCO2 an appealing methodology that can replace or diminish the exposure of harsh chemicals to sensitive materials, which in turn could lead to better preservation of their biochemical and mechanical properties. The presented review covers relevant literature over the last years where scCO2-assisted decellularization is employed, as well as discussing major topics such as the mechanism of action behind scCO2-assisted decellularization, CO2 and cosolvents' solvent properties, effect of the operational parameters on decellularization efficacy and on the material's properties.
Collapse
Affiliation(s)
- Marta M Duarte
- CBQF - Centro de Biotecnologia e Química Fina - Laboratório Associado, Universidade Católica Portuguesa, Escola Superior de Biotecnologia, Rua Diogo Botelho 1327, 4169-005 Porto, Portugal.
| | - Inês V Silva
- CBQF - Centro de Biotecnologia e Química Fina - Laboratório Associado, Universidade Católica Portuguesa, Escola Superior de Biotecnologia, Rua Diogo Botelho 1327, 4169-005 Porto, Portugal.
| | | | - Nina Bionda
- iFyber, LLC, 950 Danby Road, Ithaca, NY 14850, USA
| | - Ana Rita C Duarte
- LAQV/REQUIMTE, Departamento de Química, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, Caparica, Portugal
| | - Ana L Oliveira
- CBQF - Centro de Biotecnologia e Química Fina - Laboratório Associado, Universidade Católica Portuguesa, Escola Superior de Biotecnologia, Rua Diogo Botelho 1327, 4169-005 Porto, Portugal.
| |
Collapse
|
27
|
Wang J, Zhang Y, Sun J, Jiao Z. Controllable fabrication of multi‐modal porous
PLGA
scaffolds with different sizes of
SPIONs
using supercritical
CO
2
foaming. J Appl Polym Sci 2022. [DOI: 10.1002/app.52287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- Jinjing Wang
- School of Energy and Environment Southeast University Nanjing Jiangsu China
- Jiangsu Key Laboratory for Biomaterials and Devices Nanjing Jiangsu China
- Joint Research Institute of Southeast University and Monash University Southeast University Suzhou Jiangsu China
| | - Yi Zhang
- School of Energy and Environment Southeast University Nanjing Jiangsu China
- Jiangsu Key Laboratory for Biomaterials and Devices Nanjing Jiangsu China
- Joint Research Institute of Southeast University and Monash University Southeast University Suzhou Jiangsu China
| | - Jianfei Sun
- Jiangsu Key Laboratory for Biomaterials and Devices Nanjing Jiangsu China
- Joint Research Institute of Southeast University and Monash University Southeast University Suzhou Jiangsu China
| | - Zhen Jiao
- Jiangsu Key Laboratory for Biomaterials and Devices Nanjing Jiangsu China
- Joint Research Institute of Southeast University and Monash University Southeast University Suzhou Jiangsu China
- School of Chemistry and Chemical Engineering Southeast University Nanjing Jiangsu China
| |
Collapse
|
28
|
Hassan M, Sulaiman M, Yuvaraju PD, Galiwango E, Rehman IU, Al-Marzouqi AH, Khaleel A, Mohsin S. Biomimetic PLGA/Strontium-Zinc Nano Hydroxyapatite Composite Scaffolds for Bone Regeneration. J Funct Biomater 2022; 13:jfb13010013. [PMID: 35225976 PMCID: PMC8883951 DOI: 10.3390/jfb13010013] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 01/22/2022] [Accepted: 01/24/2022] [Indexed: 02/04/2023] Open
Abstract
Synthetic bone graft substitutes have attracted increasing attention in tissue engineering. This study aimed to fabricate a novel, bioactive, porous scaffold that can be used as a bone substitute. Strontium and zinc doped nano-hydroxyapatite (Sr/Zn n-HAp) were synthesized by a water-based sol-gel technique. Sr/Zn n-HAp and poly (lactide-co-glycolide) (PLGA) were used to fabricate composite scaffolds by supercritical carbon dioxide technique. FTIR, XRD, TEM, SEM, and TGA were used to characterize Sr/Zn n-HAp and the composite scaffolds. The synthesized scaffolds were adequately porous with an average pore size range between 189 to 406 µm. The scaffolds demonstrated bioactive behavior by forming crystals when immersed in the simulated body fluid. The scaffolds after immersing in Tris/HCl buffer increased the pH value of the medium, establishing their favorable biodegradable behavior. ICP-MS study for the scaffolds detected the presence of Sr, Ca, and Zn ions in the SBF within the first week, which would augment osseointegration if implanted in the body. nHAp and their composites (PLGA-nHAp) showed ultimate compressive strength ranging between 0.4–19.8 MPa. A 2.5% Sr/Zn substituted nHAp-PLGA composite showed a compressive behavior resembling that of cancellous bone indicating it as a good candidate for cancellous bone substitute.
Collapse
Affiliation(s)
- Mozan Hassan
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates; (M.H.); (M.S.)
| | - Mohsin Sulaiman
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates; (M.H.); (M.S.)
| | - Priya Dharshini Yuvaraju
- Department of Pharmacology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates;
| | - Emmanuel Galiwango
- Department of Chemical and Petroleum Engineering, College of Engineering, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates; (E.G.); (A.H.A.-M.)
- Energy Systems and Nuclear Science Faculty, Ontario Tech University, Oshawa, ON L1G 8C4, Canada
| | - Ihtesham ur Rehman
- Engineering Department, Faculty of Science and Technology, Lancaster University, Gillow Avenue, Lancaster LA1 4YW, UK;
| | - Ali H. Al-Marzouqi
- Department of Chemical and Petroleum Engineering, College of Engineering, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates; (E.G.); (A.H.A.-M.)
| | - Abbas Khaleel
- Department of Chemistry, College of Science, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates;
| | - Sahar Mohsin
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates; (M.H.); (M.S.)
- Correspondence: ; Tel.: +971-3-713-7516
| |
Collapse
|
29
|
Lu J, Hu D, Ma C, Xu X, Shen L, Rong J, Zhao J, Shuai B. Modified Qing' e Pills exerts anti-osteoporosis effects and prevents bone loss by enhancing type H blood vessel formation. Front Endocrinol (Lausanne) 2022; 13:998971. [PMID: 36147560 PMCID: PMC9485463 DOI: 10.3389/fendo.2022.998971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 08/18/2022] [Indexed: 11/29/2022] Open
Abstract
OBJECTIVE To explore whether the modified Qing' e Pills (MQEP) exerts anti-osteoporotic effects and prevents bone loss by enhancing angiogenesis. METHODS Network pharmacology was used to assess whether MQEP has a pro-angiogenic capacity and to predict its potential targets. Human umbilical vein endothelial cells were treated with glucocorticoids and MQEP to assess cell viability. The expression of angiotensin II type 1 receptor, angiotensin II type 2 receptor, and angiotensin converting enzyme, which are associated with the activation of the renin-angiotensin-aldosterone system, and the expression of vascular endothelial growth factor and hypoxia-inducible factor 1 alpha, which are associated with the formation of type H blood vessels, were examined by western blot and RT-qPCR. Thereafter, the glucocorticoid-induced osteoporosis model was established and intervened with MQEP. Femur scanning was performed with micro-computed tomography; trabecular spacing, trabecular thickness, and trabecular number were observed and calculated; the expression of nuclear factor-kappa B ligand and osteoprotegerin was detected by ELISA, and the ratio was calculated to evaluate the degree of bone resorption. Finally, type H blood vessels that were highly coupled to osteogenic cells were identified by immunohistochemistry staining and flow cytometry. RESULTS This is the first study to reveal and confirm that MQEP could prevent bone loss in glucocorticoid-induced osteoporosis by promoting the expression of hypoxia-inducible factor 1 alpha and vascular endothelial growth factor, which are highly associated with type H blood vessel formation. In vitro experiments confirmed that MQEP could effectively promote the proliferation of vascular endothelial cells and alleviate glucocorticoids-induced activation of the renin-angiotensin-aldosterone system, thereby reducing vascular injury. CONCLUSION MQEP exerts anti-osteoporosis effects and prevents bone loss by alleviating vascular injury caused by renin-angiotensin-aldosterone system activation and promoting type H blood vessel formation.
Collapse
Affiliation(s)
- Junjie Lu
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Desheng Hu
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chen Ma
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaojuan Xu
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lin Shen
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jianhui Rong
- School of Chinese Medicine, The University of Hong Kong, Pokfulam, Hong Kong, SAR China
| | - Jia Zhao
- School of Chinese Medicine, The University of Hong Kong, Pokfulam, Hong Kong, SAR China
| | - Bo Shuai
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- *Correspondence: Bo Shuai,
| |
Collapse
|
30
|
Lu W, Zhou C, Ma Y, Li J, Chen Y, Jiang J, Dong L, He F. Improved osseointegration of strontium-modified titanium implant by regulating angiogenesis and macrophage polarization. Biomater Sci 2022; 10:2198-2214. [DOI: 10.1039/d1bm01488a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Strotium (Sr) has shown strong osteogenic potential and thereby been widely incorporated into dental and orthopedic implants. However, the improved osseointegration of strontium-modified titanium implant through regulation of angiogenesis and macrophage...
Collapse
|
31
|
Wang L, Wang C, Zhou L, Bi Z, Shi M, Wang D, Li Q. Fabrication of a novel Three-Dimensional porous PCL/PLA tissue engineering scaffold with high connectivity for endothelial cell migration. Eur Polym J 2021. [DOI: 10.1016/j.eurpolymj.2021.110834] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
32
|
Ding A, Li CH, Yu CY, Zhou HT, Zhang ZH. Long non-coding RNA MALAT1 enhances angiogenesis during bone regeneration by regulating the miR-494/SP1 axis. J Transl Med 2021; 101:1458-1466. [PMID: 34392309 DOI: 10.1038/s41374-021-00649-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 07/21/2021] [Accepted: 07/26/2021] [Indexed: 01/16/2023] Open
Abstract
Bone regeneration is a coordinated process involving connections between blood vessels and osteocytes. Angiogenesis and osteogenesis are tightly connected throughout the progression of bone regeneration. This study aimed to explore the underlying mechanism of metastasis-associated lung adenocarcinoma transcript 1 (MALAT1)-regulated angiogenesis during bone regeneration. Gene and protein expression was detected by quantitative real-time PCR and western blot assay. Vascular endothelial growth factor (VEGFA) secretion was assessed by enzyme-linked immunosorbent assay. To evaluate the effect of osteogenic differentiation, alkaline phosphatase (ALP) and alizarin red staining assays were performed. Proliferation was detected by 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay. Migration and angiogenesis were measured using Transwell and tube formation assays. A dual luciferase reporter assay was performed to confirm the binding relationship among MALAT1, miR-494, and specificity protein 1 (SP1). Expression levels of MALAT1, SP1, and VEGFA were elevated and miR-494 was suppressed in MC3T3-E1 cells after culture in osteogenic medium. MALAT1 knockdown suppressed the osteogenic differentiation of MC3T3-E1, since ALP activity, mineralized nodules, and expression of the osteodifferentiated markers runt-related transcription factor 2 and osterix were restrained. In addition, MALAT1 silencing inhibited angiogenesis during bone regeneration, as the proliferation, migration, and capillary tube formation of human umbilical vein endothelial cells were blocked. Furthermore, miR-494 was directly targeted by MALAT1 and regulated the SP1/Toll-like receptor 2 (TLR2)/bone morphogenetic protein 2 (BMP2) axis by targeting SP1. Furthermore, miR-494 overexpression inhibited angiogenesis and osteogenic differentiation. Moreover, SP1 overexpression or miR-494 inhibition rescued the regulatory effect of sh-MALAT1 on angiogenesis and osteogenic differentiation. Taken together, these findings indicate that MALAT1 promotes angiogenesis and osteogenic differentiation by targeting miR-494 and activating the SP1/TLR2/BMP2 pathway, suggesting a novel target for bone regeneration therapy by promoting angiogenesis.
Collapse
Affiliation(s)
- Ao Ding
- Department of Stomatology, The First Affiliated Hospital of USTC (Anhui Provincial Hospital), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui Province, P.R. China
| | - Cheng-Hua Li
- Department of Stomatology, Beidaihe Rihabilitation and Recuperation Center of PLA, Qinhuangdao, Hebei Province, P.R. China
| | - Chan-Yuan Yu
- Department of Stomatology, The First Affiliated Hospital of USTC (Anhui Provincial Hospital), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui Province, P.R. China
| | - Hang-Tian Zhou
- Department of Stomatology, The First Affiliated Hospital of USTC (Anhui Provincial Hospital), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui Province, P.R. China
| | - Zhi-Hong Zhang
- Department of Stomatology, The First Affiliated Hospital of USTC (Anhui Provincial Hospital), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui Province, P.R. China.
| |
Collapse
|
33
|
Deng M, Tan J, Dai Q, Luo F, Xu J. Macrophage-Mediated Bone Formation in Scaffolds Modified With MSC-Derived Extracellular Matrix Is Dependent on the Migration Inhibitory Factor Signaling Pathway. Front Cell Dev Biol 2021; 9:714011. [PMID: 34621738 PMCID: PMC8490662 DOI: 10.3389/fcell.2021.714011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 08/09/2021] [Indexed: 12/25/2022] Open
Abstract
The positive role of macrophages in the osteogenesis of mesenchymal stem cells (MSCs) has been a recent research focus. On the other hand, MSCs could carefully regulate the paracrine molecules derived from macrophages. Human umbilical cord mesenchymal stem cells (hucMSCs) can reduce the secretion of inflammatory factors from macrophages to improve injury healing. hucMSC-derived extracellular matrix (hucMSC-ECM) has the similar effect to hucMSCs, which could combat the inflammatory response of macrophages. Additionally, MSC-derived extracellular matrix also enhanced bone regeneration by inhibiting osteoclastic differentiation of monocyte/macrophage lineage. However, whether hucMSC-ECM could improve bone formation by guiding macrophage-induced osteogenic differentiation of MSCs is unknown. Here, we present decalcified bone scaffolds modified by hucMSC-derived extracellular matrix (DBM-ECM), which maintained multiple soluble cytokines from hucMSCs, including macrophage migration inhibitory factor (MIF). Compared with DBM, the DBM-ECM scaffolds induced bone formation in an improved heterotopic ossification model of severe combined immunodeficiency (SCID) mice in a macrophage-dependent manner. Macrophages cocultured with DBM-ECM expressed four osteoinductive cytokines (BMP2, FGF2, TGFβ3 and OSM), which were screened out by RNA sequencing and measured by qPCR and western blot. The conditioned medium from macrophages cocultured with DBM-ECM improved the osteogenic differentiation of hBMSCs. Furthermore, DBM-ECM activated CD74/CD44 (the typical MIF receptors) signal transduction in macrophages, including phosphorylation of P38 and dephosphorylation of c-jun. On the other side, the inhibitory effects of the DBM-ECM scaffolds with a deficient of MIF on osteogenesis in vitro and in vivo revealed that macrophage-mediated osteogenesis depended on MIF/CD74 signal transduction. The results of this study indicate that the coordinated crosstalk of macrophages and MSCs plays a key role on bone regeneration, with an emphasis on hucMSC-ECM constructing a macrophage-derived osteoinductive microenvironment.
Collapse
Affiliation(s)
- Moyuan Deng
- Department of Orthopaedics, Southwest Hospital, Army Medical University, Chongqing, China
| | - Jiulin Tan
- Department of Orthopaedics, Southwest Hospital, Army Medical University, Chongqing, China
| | - Qijie Dai
- Department of Orthopaedics, Southwest Hospital, Army Medical University, Chongqing, China
| | - Fei Luo
- Department of Orthopaedics, Southwest Hospital, Army Medical University, Chongqing, China
| | - Jianzhong Xu
- Department of Orthopaedics, Southwest Hospital, Army Medical University, Chongqing, China
| |
Collapse
|
34
|
Xu HZ, Su JS. Restoration of critical defects in the rabbit mandible using osteoblasts and vascular endothelial cells co-cultured with vascular stent-loaded nano-composite scaffolds. J Mech Behav Biomed Mater 2021; 124:104831. [PMID: 34555626 DOI: 10.1016/j.jmbbm.2021.104831] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 09/04/2021] [Accepted: 09/07/2021] [Indexed: 01/07/2023]
Abstract
The success of large bone defect repair with tissue engineering technology depends mainly on angiogenesis and osteogenesis. In this study, we prepared poly-caprolactone/nano-hydroxyapatite/beta-calcium phosphate (PCL/nHA/β-TCP) composite scaffolds loaded with poly-(lactic-co-glycolic acid)/nano-hydroxyapatite/collagen/heparin sodium (PLGA/nHA/Col/HS) nanofiber small vascular stent by electrospinning and hot press forming-particle leaching methods. Supramolecular electrostatic self-assembly technology was used to modify the surfaces of small vascular stents to aid in hydrophilicity and anticoagulation. The surfaces of composite scaffolds were modified with an Arg-Gly-Asp (RGD) short peptide by physical adsorption to supply cell adhesion sites. The scaffolds were then combined with rabbit bone marrow-derived osteoblasts (OBs) and rabbit bone marrow-derived vascular endothelial cells (RVECs) to construct large, biologically active vascularized tissue-engineered bone in vitro; this bone was then used to repair critical bone defects in rabbit mandibles. Mechanical and biocompatibility testing results showed that PCL/nHA/β-TCP composite scaffolds loaded with small vascular stents had good surface structure, mechanical properties, biocompatibility, and bone-regeneration induction potential. Twelve weeks after implantation, histological analysis and X-ray scans showed that the use of osteoblasts and vascular endothelial cells co-cultured with PCL/nHA/β-TCP scaffolds was sufficient to repair critical defects in rabbit mandibles.
Collapse
Affiliation(s)
- Hong Zhen Xu
- Department of Prosthodontics, Shanghai Stomatological Hospital, Fudan University, Shanghai, China
| | - Jian Sheng Su
- Department of Prosthodontics, School & Hospital of Stomatology, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Tongji University, Shanghai, China.
| |
Collapse
|
35
|
Jin S, Gao J, Yang R, Yuan C, Wang R, Zou Q, Zuo Y, Zhu M, Li Y, Man Y, Li J. A baicalin-loaded coaxial nanofiber scaffold regulated inflammation and osteoclast differentiation for vascularized bone regeneration. Bioact Mater 2021; 8:559-572. [PMID: 34541420 PMCID: PMC8436066 DOI: 10.1016/j.bioactmat.2021.06.028] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Revised: 06/01/2021] [Accepted: 06/23/2021] [Indexed: 02/05/2023] Open
Abstract
We demonstrate a simple, effective and feasible method to address the shrinkage of Poly (lactic-co-glycolic acid) (PLGA) through a core-shell structure fiber strategy. The results revealed that introducing size-stable poly-caprolactone (PCL) as the core fiber significantly improved the PLGA-based fibrous scaffold's dimensional maintenance. We further utilized fish collagen to modify the PLGA shell layer (PFC) of coaxial fibers and loaded baicalin (BA) into the PCL core layer (PCL-BA) to endow fibrous scaffold with more functional biological cues. The PFC/PCL-BA fibrous scaffold promoted the osteogenic differentiation of bone mesenchymal stem cells and stimulated the RAW264.7 cells to polarize into a pro-reparative phenotype. Importantly, the in vivo study demonstrated that the PFC/PCL-BA scaffold could regulate inflammation and osteoclast differentiation, favor neovascularization and bone formation. This work tactfully combined PLGA and PCL to establish a drug release platform based on the core-shell fibrous scaffold for vascularized bone regeneration. A multifunctional baicalin-loaded coaxial fiber scaffold prepared by electrospinning. The coaxial nanofiber can effectively resist the shrinkage of PLGA. Baicalin endow the nanofibrous scaffold with excellent biological properties. The scaffold can alleviate the inflammation and achieve vascularized bone regeneration.
Collapse
Affiliation(s)
- Shue Jin
- Research Center for Nano-Biomaterials, Analytical and Testing Center, Sichuan University, Chengdu, 610064, PR China
| | - Jing Gao
- Research Center for Nano-Biomaterials, Analytical and Testing Center, Sichuan University, Chengdu, 610064, PR China
| | - Renli Yang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610065, PR China
| | - Chen Yuan
- Research Center for Nano-Biomaterials, Analytical and Testing Center, Sichuan University, Chengdu, 610064, PR China
| | - Ruili Wang
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Materials Science and Engineering, Donghua University, Shanghai, 201620, PR China
| | - Qin Zou
- Research Center for Nano-Biomaterials, Analytical and Testing Center, Sichuan University, Chengdu, 610064, PR China
| | - Yi Zuo
- Research Center for Nano-Biomaterials, Analytical and Testing Center, Sichuan University, Chengdu, 610064, PR China
| | - Meifang Zhu
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Materials Science and Engineering, Donghua University, Shanghai, 201620, PR China
| | - Yubao Li
- Research Center for Nano-Biomaterials, Analytical and Testing Center, Sichuan University, Chengdu, 610064, PR China
| | - Yi Man
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610065, PR China
| | - Jidong Li
- Research Center for Nano-Biomaterials, Analytical and Testing Center, Sichuan University, Chengdu, 610064, PR China
| |
Collapse
|
36
|
Chen MH, Wang YH, Sun BJ, Yu LM, Chen QQ, Han XX, Liu YH. HIF-1α activator DMOG inhibits alveolar bone resorption in murine periodontitis by regulating macrophage polarization. Int Immunopharmacol 2021; 99:107901. [PMID: 34273637 DOI: 10.1016/j.intimp.2021.107901] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 02/10/2021] [Accepted: 06/16/2021] [Indexed: 11/24/2022]
Abstract
Periodontitis is initiated by serious and sustained bacterial infection and ultimately results in chronic immune-mediated inflammation, tissue destruction, and bone loss. The pathogenesis of periodontitis remains unclear. Host immunological responses to periodontal bacteria ultimately determine the severity and mechanisms governing periodontitis progression. This study aimed to clarify the effect of the hypoxia-inducible factor-1α (HIF-1α) activator dimethyloxalylglycine (DMOG) on a mouse periodontitis model and its underlying role in macrophage polarization. qRT-PCR analysis showed that DMOG inhibited the M1-like polarization of both RAW264.7 macrophages and murine bone marrow macrophages (BMMs) and downregulated TNF-α, IL-6, CD86, and MCP-1 expression in vitro. Immunofluorescence staining and flow cytometry also confirmed the less percentage of F4/80 + CD86 + cells after DMOG treatment. The phosphorylation of NF-κB pathway was also inhibited by DMOG with higher level of HIF-1α expression. Furthermore, mice treated with DMOG showed decreased alveolar bone resorption in the experimental periodontitis model, with significant increases in alveolar bone volume/tissue volume (BV/TV) and bone mineral density (BMD). DMOG treatment of mice decreased the ratio of M1/M2 (CD86+/CD206+) macrophages in periodontal tissues, resulting in the downregulation of proinflammatory cytokines such as TNF-α and IL-6 and increased levels of anti-inflammatory factors such as IL-4 and IL-10. DMOG treatment promoted the number of HIF-1α-positive cells in periodontal tissues. This study demonstrated the cell-specific roles of DMOG in macrophage polarization in vitro and provided insight into the mechanism underlying the protective effect of DMOG in a model of periodontitis.
Collapse
Affiliation(s)
- Mei-Hua Chen
- Department of Periodontology, Shanghai Stomatological Hospital, Fudan University, Shanghai, China; Oral Biomedical Engineering Laboratory, Shanghai Stomatological Hospital, Fudan University, Shanghai, China
| | - Yu-Hui Wang
- Oral Biomedical Engineering Laboratory, Shanghai Stomatological Hospital, Fudan University, Shanghai, China; Department of Orthodontics, Shanghai Stomatological Hospital, Fudan University, Shanghai, China
| | - Bing-Jing Sun
- Oral Biomedical Engineering Laboratory, Shanghai Stomatological Hospital, Fudan University, Shanghai, China; Department of Orthodontics, Shanghai Stomatological Hospital, Fudan University, Shanghai, China
| | - Li-Ming Yu
- Oral Biomedical Engineering Laboratory, Shanghai Stomatological Hospital, Fudan University, Shanghai, China; Department of Orthodontics, Shanghai Stomatological Hospital, Fudan University, Shanghai, China
| | - Qing-Qing Chen
- Oral Biomedical Engineering Laboratory, Shanghai Stomatological Hospital, Fudan University, Shanghai, China; Department of Orthodontics, Shanghai Stomatological Hospital, Fudan University, Shanghai, China
| | - Xin-Xin Han
- Oral Biomedical Engineering Laboratory, Shanghai Stomatological Hospital, Fudan University, Shanghai, China
| | - Yue-Hua Liu
- Oral Biomedical Engineering Laboratory, Shanghai Stomatological Hospital, Fudan University, Shanghai, China; Department of Orthodontics, Shanghai Stomatological Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
37
|
Determining the Optimal Conditions for the Production by Supercritical CO 2 of Biodegradable PLGA Foams for the Controlled Release of Rutin as a Medical Treatment. Polymers (Basel) 2021; 13:polym13101645. [PMID: 34069337 PMCID: PMC8158779 DOI: 10.3390/polym13101645] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 05/14/2021] [Accepted: 05/14/2021] [Indexed: 01/09/2023] Open
Abstract
Poly(D,L,-lactide-co-glycolide) (PLGA) foam samples impregnated with rutin were successfully produced by supercritical foaming processes. A number of parameters such as pressure (80–200 bar), temperature (35–55 °C), depressurization rate (5–100 bar/min), ratio lactide:glycolide of the poly(D,L,-lactide-co-glycolide) (50:50 and 75:25) were studied to determine their effect on the expansion factor and on the glass transition temperature of the polymer foams and their consequences on the release profile of the rutin entrapped in them. The impregnated foams were characterized by scanning electron microscopy, differential scanning calorimetry, and mercury intrusion porosimetry. A greater impregnation of rutin into the polymer foam pores was observed as pressure was increased. The release of rutin in a phosphate buffer solution was investigated. The controlled release tests confirmed that the modification of certain variables would result in considerable differences in the drug release profiles. Thus, five-day drug release periods were achieved under high pressure and temperature while the depressurization rate remained low.
Collapse
|
38
|
Fu L, Zhang L, Zhang X, Chen L, Cai Q, Yang X. Roles of oxygen level and hypoxia-inducible factor signaling pathway in cartilage, bone and osteochondral tissue engineering. Biomed Mater 2021; 16:022006. [PMID: 33440367 DOI: 10.1088/1748-605x/abdb73] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The repair and treatment of articular cartilage injury is a huge challenge of orthopedics. Currently, most of the clinical methods applied in treating cartilage injuries are mainly to relieve pains rather than to cure them, while the strategy of tissue engineering is highly expected to achieve the successful repair of osteochondral defects. Clear understandings of the physiological structures and mechanical properties of cartilage, bone and osteochondral tissues have been established, but the understanding of their physiological heterogeneity still needs further investigation. Apart from the gradients in the micromorphology and composition of cartilage-to-bone extracellular matrixes, an oxygen gradient also exists in natural osteochondral tissue. The response of hypoxia-inducible factor (HIF)-mediated cells to oxygen would affect the differentiation of stem cells and the maturation of osteochondral tissue. This article reviews the roles of oxygen level and HIF signaling pathway in the development of articular cartilage tissue, and their prospective applications in bone and cartilage tissue engineering. The strategies for regulating HIF signaling pathway and how these strategies finding their potential applications in the regeneration of integrated osteochondral tissue are also discussed.
Collapse
Affiliation(s)
- Lei Fu
- State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029, People's Republic of China
| | | | | | | | | | | |
Collapse
|
39
|
Salètes M, Vartin M, Mocquot C, Chevalier C, Grosgogeat B, Colon P, Attik N. Mesoporous Bioactive Glasses Cytocompatibility Assessment: A Review of In Vitro Studies. Biomimetics (Basel) 2021; 6:9. [PMID: 33498616 PMCID: PMC7839003 DOI: 10.3390/biomimetics6010009] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 01/11/2021] [Accepted: 01/20/2021] [Indexed: 12/12/2022] Open
Abstract
Thanks to their high porosity and surface area, mesoporous bioactive glasses (MBGs) have gained significant interest in the field of medical applications, in particular, with regards to enhanced bioactive properties which facilitate bone regeneration. The aim of this article is to review the state of the art regarding the biocompatibility evaluation of MBGs and provide a discussion of the various approaches taken. The research was performed using PubMed database and covered articles published in the last five years. From a total of 91 articles, 63 were selected after analyzing them according to our inclusion and exclusion criteria. In vitro methodologies and techniques used for biocompatibility assessment were investigated. Among the biocompatibility assessment techniques, scanning electron microscopy (SEM) has been widely used to study cell morphology and adhesion. Viability and proliferation were assessed using different assays including cell counting and/or cell metabolic activity measurement. Finally, cell differentiation tests relied on the alkaline phosphatase assay; however, these were often complemented by specific bimolecular tests according to the exact application of the mesoporous bioactive glass. The standardization and validation of all tests performed for MBG cytocompatibility is a key aspect and crucial point and should be considered in order to avoid inconsistencies, bias between studies, and unnecessary consumption of time. Therefore, introducing standard tests would serve an important role in the future assessment and development of MBG materials.
Collapse
Affiliation(s)
- Margaux Salètes
- CPE Lyon, Université Claude Bernard Lyon 1, CEDEX 08, 69372 Lyon, France; (M.S.); (M.V.)
- Laboratoire des Multimatériaux et Interfaces, UMR CNRS 5615, Université de Lyon—Université Claude Bernard Lyon 1, CEDEX 08, 69372 Lyon, France; (C.M.); (C.C.); (B.G.); (P.C.)
| | - Marta Vartin
- CPE Lyon, Université Claude Bernard Lyon 1, CEDEX 08, 69372 Lyon, France; (M.S.); (M.V.)
- Laboratoire des Multimatériaux et Interfaces, UMR CNRS 5615, Université de Lyon—Université Claude Bernard Lyon 1, CEDEX 08, 69372 Lyon, France; (C.M.); (C.C.); (B.G.); (P.C.)
| | - Caroline Mocquot
- Laboratoire des Multimatériaux et Interfaces, UMR CNRS 5615, Université de Lyon—Université Claude Bernard Lyon 1, CEDEX 08, 69372 Lyon, France; (C.M.); (C.C.); (B.G.); (P.C.)
- Assistance Publique-Hôpitaux de Paris, Hôpital Rothschild, Service D’odontologie, Faculté Dentaire, Université de Paris, 75012 Paris, France
| | - Charlène Chevalier
- Laboratoire des Multimatériaux et Interfaces, UMR CNRS 5615, Université de Lyon—Université Claude Bernard Lyon 1, CEDEX 08, 69372 Lyon, France; (C.M.); (C.C.); (B.G.); (P.C.)
| | - Brigitte Grosgogeat
- Laboratoire des Multimatériaux et Interfaces, UMR CNRS 5615, Université de Lyon—Université Claude Bernard Lyon 1, CEDEX 08, 69372 Lyon, France; (C.M.); (C.C.); (B.G.); (P.C.)
- Faculté d’Odontologie, Université de Lyon, Université Claude Bernard Lyon 1, 69008 Lyon, France
- Hospices Civils de Lyon, Service D’odontologie, 69007 Lyon, France
| | - Pierre Colon
- Laboratoire des Multimatériaux et Interfaces, UMR CNRS 5615, Université de Lyon—Université Claude Bernard Lyon 1, CEDEX 08, 69372 Lyon, France; (C.M.); (C.C.); (B.G.); (P.C.)
- Assistance Publique-Hôpitaux de Paris, Hôpital Rothschild, Service D’odontologie, Faculté Dentaire, Université de Paris, 75012 Paris, France
| | - Nina Attik
- Laboratoire des Multimatériaux et Interfaces, UMR CNRS 5615, Université de Lyon—Université Claude Bernard Lyon 1, CEDEX 08, 69372 Lyon, France; (C.M.); (C.C.); (B.G.); (P.C.)
- Faculté d’Odontologie, Université de Lyon, Université Claude Bernard Lyon 1, 69008 Lyon, France
| |
Collapse
|
40
|
Chen J, Hendriks M, Chatzis A, Ramasamy SK, Kusumbe AP. Bone Vasculature and Bone Marrow Vascular Niches in Health and Disease. J Bone Miner Res 2020; 35:2103-2120. [PMID: 32845550 DOI: 10.1002/jbmr.4171] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 07/21/2020] [Accepted: 08/05/2020] [Indexed: 12/20/2022]
Abstract
Bone vasculature and bone marrow vascular niches supply oxygen, nutrients, and secrete angiocrine factors required for the survival, maintenance, and self-renewal of stem and progenitor cells. In the skeletal system, vasculature creates nurturing niches for bone and blood-forming stem cells. Blood vessels regulate hematopoiesis and drive bone formation during development, repair, and regeneration. Dysfunctional vascular niches induce skeletal aging, bone diseases, and hematological disorders. Recent cellular and molecular characterization of the bone marrow microenvironment has provided unprecedented insights into the complexity, heterogeneity, and functions of the bone vasculature and vascular niches. The bone vasculature is composed of distinct vessel subtypes that differentially regulate osteogenesis, hematopoiesis, and disease conditions in bones. Further, bone marrow vascular niches supporting stem cells are often complex microenvironments involving multiple different cell populations and vessel subtypes. This review provides an overview of the emerging vascular cell heterogeneity in bone and the new roles of the bone vasculature and associated vascular niches in health and disease. © 2020 The Authors. Journal of Bone and Mineral Research published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Junyu Chen
- Tissue and Tumor Microenvironments Group, The Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Michelle Hendriks
- Institute of Clinical Sciences, Imperial College London, London, UK
- MRC London Institute of Medical Sciences, Imperial College London, London, UK
| | - Alexandros Chatzis
- Tissue and Tumor Microenvironments Group, The Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Saravana K Ramasamy
- Institute of Clinical Sciences, Imperial College London, London, UK
- MRC London Institute of Medical Sciences, Imperial College London, London, UK
| | - Anjali P Kusumbe
- Tissue and Tumor Microenvironments Group, The Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| |
Collapse
|
41
|
Pan Y, Zhao Y, Kuang R, Liu H, Sun D, Mao T, Jiang K, Yang X, Watanabe N, Mayo KH, Lin Q, Li J. Injectable hydrogel-loaded nano-hydroxyapatite that improves bone regeneration and alveolar ridge promotion. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 116:111158. [DOI: 10.1016/j.msec.2020.111158] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Revised: 05/29/2020] [Accepted: 06/02/2020] [Indexed: 12/20/2022]
|
42
|
任 思, 刘 艺, 朱 彦, 王 莹, 刘 鳗, 周 延. [Application status of hypoxia mimetic agents in bone tissue engineering]. ZHONGGUO XIU FU CHONG JIAN WAI KE ZA ZHI = ZHONGGUO XIUFU CHONGJIAN WAIKE ZAZHI = CHINESE JOURNAL OF REPARATIVE AND RECONSTRUCTIVE SURGERY 2020; 34:1190-1194. [PMID: 32929915 PMCID: PMC8171726 DOI: 10.7507/1002-1892.201911144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 03/30/2020] [Indexed: 11/03/2022]
Abstract
OBJECTIVE To summarize the application status of hypoxia mimetic agents in bone tissue engineering. METHODS The related literature about the hypoxia mimetic agents in bone tissue engineering was reviewed and analyzed. And the application status and progress of hypoxia mimetic agents in bone tissue engineering were retrospectively analyzed. RESULTS Hypoxia mimetic agents have the same effect as hypoxia in up-regulating the level of hypoxia inducible factor 1α (HIF-1α). The combination of hypoxia mimetic agents and scaffolds can up-regulate the level of HIF-1α in bone tissue engineering, thus promoting early vascularization and bone regeneration of the bone defect area, which provides a new idea for using bone tissue engineering to repair bone defect. At present, the commonly used hypoxia mimetic agents include iron chelating agents, oxoglutarate competitive analogues, proline hydroxylase inhibitors, etc. CONCLUSION Hypoxia mimetic agents have a wide application prospect in bone tissue engineering, but they have been used in bone tissue engineering for a short time, more attention should be paid to their possible side effects. In the future research, the hypoxia mimetic agents should be developed in the direction of higher targeting specificity and safety, and the exact mechanism of hypoxia mimetic agents in promoting bone regeneration should be further explored.
Collapse
Affiliation(s)
- 思聪 任
- 吉林大学口腔医院种植科(长春 130021)Department of Oral Implantology, Hospital of Stomatology, Jilin University, Changchun Jilin, 130021, P.R.China
| | - 艺萍 刘
- 吉林大学口腔医院种植科(长春 130021)Department of Oral Implantology, Hospital of Stomatology, Jilin University, Changchun Jilin, 130021, P.R.China
| | - 彦霖 朱
- 吉林大学口腔医院种植科(长春 130021)Department of Oral Implantology, Hospital of Stomatology, Jilin University, Changchun Jilin, 130021, P.R.China
| | - 莹莹 王
- 吉林大学口腔医院种植科(长春 130021)Department of Oral Implantology, Hospital of Stomatology, Jilin University, Changchun Jilin, 130021, P.R.China
| | - 鳗萱 刘
- 吉林大学口腔医院种植科(长春 130021)Department of Oral Implantology, Hospital of Stomatology, Jilin University, Changchun Jilin, 130021, P.R.China
| | - 延民 周
- 吉林大学口腔医院种植科(长春 130021)Department of Oral Implantology, Hospital of Stomatology, Jilin University, Changchun Jilin, 130021, P.R.China
| |
Collapse
|
43
|
Vila-Parrondo C, García-Astrain C, Liz-Marzán LM. Colloidal systems toward 3D cell culture scaffolds. Adv Colloid Interface Sci 2020; 283:102237. [PMID: 32823220 DOI: 10.1016/j.cis.2020.102237] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 08/07/2020] [Accepted: 08/07/2020] [Indexed: 12/14/2022]
Abstract
Three-dimensional porous scaffolds are essential for the development of tissue engineering and regeneration, as biomimetic supports to recreate the microenvironment present in natural tissues. To successfully achieve the growth and development of a specific kind of tissue, porous matrices should be able to influence cell behavior by promoting close cell-cell and cell-matrix interactions. To achieve this goal, the scaffold must fulfil a set of conditions, including ordered interconnected porosity to promote cell diffusion and vascularization, mechanical strength to support the tissue during continuous ingrowth, and biocompatibility to avoid toxicity. Among various building approaches to the construction of porous matrices, selected strategies afford hierarchical scaffolds with such defined properties. The control over porosity, microstructure or morphology, is crucial to the fabrication of high-end, reproducible scaffolds for the target application. In this review, we provide an insight into recent advances toward the colloidal fabrication of hierarchical scaffolds. After identifying the main requirements for scaffolds in biomedical applications, conceptual building processes are introduced. Examples of tissue regeneration applications are provided for different scaffold types, highlighting their versatility and biocompatibility. We finally provide a prospect about the current state of the art and limitations of porous scaffolds, along with challenges that are to be addressed, so these materials consolidate in the fields of tissue engineering and drug delivery.
Collapse
|
44
|
Zhang J, Pan J, Jing W. Motivating role of type H vessels in bone regeneration. Cell Prolif 2020; 53:e12874. [PMID: 33448495 PMCID: PMC7507571 DOI: 10.1111/cpr.12874] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 06/03/2020] [Accepted: 06/22/2020] [Indexed: 02/06/2023] Open
Abstract
Coupling between angiogenesis and osteogenesis has an important role in both normal bone injury repair and successful application of tissue‐engineered bone for bone defect repair. Type H blood vessels are specialized microvascular components that are closely related to the speed of bone healing. Interactions between type H endothelial cells and osteoblasts, and high expression of CD31 and EMCN render the environment surrounding these blood vessels rich in factors conducive to osteogenesis and promote the coupling of angiogenesis and osteogenesis. Type H vessels are mainly distributed in the metaphysis of bone and densely surrounded by Runx2+ and Osterix+ osteoprogenitors. Several other factors, including hypoxia‐inducible factor‐1α, Notch, platelet‐derived growth factor type BB, and slit guidance ligand 3 are involved in the coupling of type H vessel formation and osteogenesis. In this review, we summarize the identification and distribution of type H vessels and describe the mechanism for type H vessel‐mediated modulation of osteogenesis. Type H vessels provide new insights for detection of the molecular and cellular mechanisms that underlie the crosstalk between angiogenesis and osteogenesis. As a result, more feasible therapeutic approaches for treatment of bone defects by targeting type H vessels may be applied in the future.
Collapse
Affiliation(s)
- Jiankang Zhang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jian Pan
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Wei Jing
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
45
|
Bone regenerative potential of the selective sphingosine 1-phosphate receptor modulator siponimod: In vitro characterisation using osteoblast and endothelial cells. Eur J Pharmacol 2020; 882:173262. [PMID: 32534075 DOI: 10.1016/j.ejphar.2020.173262] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 06/02/2020] [Accepted: 06/08/2020] [Indexed: 12/31/2022]
Abstract
The repair of critical bone defects remains a significant therapeutic challenge. While the implantation of drug-eluting scaffolds is an option, a drug with the optimal pharmacological properties has not yet been identified. Agents acting at sphingosine 1-phosphate (S1P) receptors have been considered, but those investigated so far do not discriminate between the five known S1P receptors. This work was undertaken to investigate the potential of the specific S1P1/5 modulator siponimod as a bone regenerative agent, by testing in vitro its effect on cell types critical to the bone regeneration process. hFOB osteoblasts and HUVEC endothelial cells were treated with siponimod and other S1P receptor modulators and investigated for changes in intracellular cyclic AMP content, viability, proliferation, differentiation, attachment and cellular motility. Siponimod showed no effect on the viability and proliferation of osteoblasts and endothelial cells, but increased osteoblast differentiation (as shown by increased alkaline phosphatase activity). Furthermore, siponimod significantly increased endothelial cell motility in scratch and transwell migration assays. These effects on osteoblast differentiation and endothelial cell migration suggest that siponimod may be a potential agent for the stimulation of localised differentiation of osteoblasts in critical bone defects.
Collapse
|
46
|
Ranmuthu CDS, Ranmuthu CKI, Russell JC, Singhania D, Khan WS. Evaluating the Effect of Non-cellular Bioactive Glass-Containing Scaffolds on Osteogenesis and Angiogenesis in in vivo Animal Bone Defect Models. Front Bioeng Biotechnol 2020; 8:430. [PMID: 32478053 PMCID: PMC7240009 DOI: 10.3389/fbioe.2020.00430] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 04/15/2020] [Indexed: 12/12/2022] Open
Abstract
The use of bone scaffolds to replace injured or diseased bone has many advantages over the currently used autologous and allogeneic options in clinical practice. This systematic review evaluates the current evidence for non-cellular scaffolds containing bioactive glass on osteogenesis and angiogenesis in animal bone defect models. Studies that reported results of osteogenesis via micro-CT and results of angiogenesis via Microfil perfusion or immunohistochemistry were included in the review. A literature search of PubMed, EMBASE and Scopus was carried out in November 2019 from which nine studies met the inclusion and exclusion criteria. Despite the significant heterogeneity in the composition of the scaffolds used in each study, it could be concluded that scaffolds containing bioactive glass improve bone regeneration in these models, both by osteogenic and angiogenic measures. Incorporation of additional elements into the glass network, using additives, and using biochemical factors generally had a beneficial effect. Comparing the different compositions of non-cellular bioactive glass containing scaffolds is however difficult due to the heterogeneity in bioactive glass compositions, fabrication methods and biochemical additives used.
Collapse
Affiliation(s)
| | | | - Jodie C. Russell
- Cambridge Clinical School, University of Cambridge, Cambridge, United Kingdom
| | - Disha Singhania
- Cambridge Clinical School, University of Cambridge, Cambridge, United Kingdom
| | - Wasim S. Khan
- Division of Trauma and Orthopaedics, Department of Surgery, Addenbrooke's Hospital, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
47
|
Liu Y, Yang S, Cao L, Zhang X, Wang J, Liu C. Facilitated vascularization and enhanced bone regeneration by manipulation hierarchical pore structure of scaffolds. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 110:110622. [PMID: 32204064 DOI: 10.1016/j.msec.2019.110622] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 12/29/2019] [Accepted: 12/31/2019] [Indexed: 12/31/2022]
Abstract
Sufficient vascularization is quite important for preventing cell death and promoting host integration during the repair of the critical sized bone defects. Porous structure providing enough space for the ingrowth of vessels is an essential consideration during the scaffold's development. In this study, we designed and fabricated three kinds of porous structured scaffolds based on poly(3-hydroxybutyrate-co-3-hydroxyhexanoate) (PHBHHx), such as mono-structured PHBHHx scaffolds with macro pores (PH-1), di-structured PHBHHx scaffolds with macro-meso pores (PHS-2), and tri-structured PHBHHx scaffolds with macro-micro-meso pores (PHS-3), respectively. In vitro effects of the hierarchical porous scaffolds on human umbilical vein endothelial cells (HUVECs), such as cell attachment, glucose and lactate detection, relative gene expressions of endothelial markers were investigated. The PHS-3 scaffolds exhibited preferential potency of inducing better angiogenesis in vitro. Consequently, the hierarchical porous scaffolds were applied to load rhBMP-2 and repair the critical sized bone defect (15 mm) in rabbits. Microangiography analysis by three dimensional micro-computed tomographic (micro-CT) demonstrated that the volume of blood vessels within the defect area was higher in the rhBMP-2 loaded PHS-3 (PHS-3/rhBMP-2) than that in other rhBMP-2 loaded porous scaffolds with simplex or double scaled pores (PH-1/rhBMP-2 or PHS-2/rhBMP-2) at 4 weeks and 8 weeks, which implied that multi-level porous structure was conducive to nutrition transmission and revascularization. Further investigations of orthotopic bone formation by micro-CT, histological and immunohistochemistry analysis confirmed the most accelerated new bone formation rate in the PHS-3/rhBMP-2 group. The maximum load value of the regenerated bone induced by PHS-3/rhBMP-2 at 12 weeks was 258.47 ± 14.77 N which did not show significant difference from the normal bone of 268.81 ± 12.05 N. These results highlighted that introducing multi-level pores into the biocompatible scaffolds may be an effective approach to promote angiogenesis and bone regeneration.
Collapse
Affiliation(s)
- Yang Liu
- The State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, People's Republic of China; Engineering Research Center for Biomedical Materials of Ministry of Education, East China University of Science and Technology, Shanghai 200237, People's Republic of China
| | - Shengbing Yang
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai 200011, People's Republic of China
| | - Lingyan Cao
- Department of Prosthodontics, Oral Bioengineering and Regenerative Medicine Lab, Shanghai Key Laboratory of Stomatology, Ninth People's Hospital Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai 200011, People's Republic of China
| | - Xiaohui Zhang
- The State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, People's Republic of China; Engineering Research Center for Biomedical Materials of Ministry of Education, East China University of Science and Technology, Shanghai 200237, People's Republic of China
| | - Jing Wang
- The State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, People's Republic of China; Engineering Research Center for Biomedical Materials of Ministry of Education, East China University of Science and Technology, Shanghai 200237, People's Republic of China.
| | - Changsheng Liu
- Engineering Research Center for Biomedical Materials of Ministry of Education, East China University of Science and Technology, Shanghai 200237, People's Republic of China; Key Laboratory for Ultrafine Materials of Ministry of Education, East China University of Science and Technology, Shanghai 200237, People's Republic of China.
| |
Collapse
|
48
|
Hou J, Jiang J, Guo H, Guo X, Wang X, Shen Y, Li Q. Fabrication of fibrillated and interconnected porous poly(ε-caprolactone) vascular tissue engineering scaffolds by microcellular foaming and polymer leaching. RSC Adv 2020; 10:10055-10066. [PMID: 35498611 PMCID: PMC9050225 DOI: 10.1039/d0ra00956c] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 03/01/2020] [Indexed: 11/21/2022] Open
Abstract
This paper provides a method combining eco-friendly supercritical CO2 microcellular foaming and polymer leaching to fabricate small-diameter vascular tissue engineering scaffolds.
Collapse
Affiliation(s)
- Jianhua Hou
- School of Mechanics & Engineering Science
- Zhengzhou University
- National Center for International Joint Research of Micro-Nano Molding Technology
- Zhengzhou
- PR China
| | - Jing Jiang
- School of Mechanics & Engineering Science
- Zhengzhou University
- National Center for International Joint Research of Micro-Nano Molding Technology
- Zhengzhou
- PR China
| | - Haiyang Guo
- School of Mechanics & Engineering Science
- Zhengzhou University
- National Center for International Joint Research of Micro-Nano Molding Technology
- Zhengzhou
- PR China
| | - Xin Guo
- School of Mechanics & Engineering Science
- Zhengzhou University
- National Center for International Joint Research of Micro-Nano Molding Technology
- Zhengzhou
- PR China
| | - Xiaofeng Wang
- School of Mechanics & Engineering Science
- Zhengzhou University
- National Center for International Joint Research of Micro-Nano Molding Technology
- Zhengzhou
- PR China
| | - Yaqiang Shen
- Shenzhen ZhaoWei Machinery & Electronics Co.,Ltd
- Shenzhen
- PR China
| | - Qian Li
- School of Mechanics & Engineering Science
- Zhengzhou University
- National Center for International Joint Research of Micro-Nano Molding Technology
- Zhengzhou
- PR China
| |
Collapse
|