1
|
Yang Q, Xu M, Fang H, Gao Y, Zhu D, Wang J, Chen Y. Targeting micromotion for mimicking natural bone healing by using NIPAM/Nb 2C hydrogel. Bioact Mater 2024; 39:41-58. [PMID: 38800718 PMCID: PMC11127186 DOI: 10.1016/j.bioactmat.2024.05.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 05/09/2024] [Accepted: 05/10/2024] [Indexed: 05/29/2024] Open
Abstract
Natural fracture healing is most efficient when the fine-tuned mechanical force and proper micromotion are applied. To mimick this micromotion at the fracture gap, a near-infrared-II (NIR-II)-activated hydrogel was fabricated by integrating two-dimensional (2D) monolayer Nb2C nanosheets into a thermally responsive poly(N-isopropylacrylamide) (NIPAM) hydrogel system. NIR-II-triggered deformation of the NIPAM/Nb2C hydrogel was designed to generate precise micromotion for co-culturing cells. It was validated that micromotion at 1/300 Hz, triggering a 2.37-fold change in the cell length/diameter ratio, is the most favorable condition for the osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs). Moreover, mRNA sequencing and verification revealed that micromotion-induced augmentation was mediated by Piezo1 activation. Suppression of Piezo1 interrupts the mechano-sensitivity and abrogates osteogenic differentiation. Calvarial and femoral shaft defect models were established to explore the biocompatibility and osteoinductivity of the Micromotion Biomaterial. A series of research methods, including radiography, micro-CT scanning, and immunohistochemical staining have been performed to evaluate biosafety and osteogenic efficacy. The in vivo results revealed that tunable micromotion strengthens the natural fracture healing process through the sequential activation of endochondral ossification, promotion of neovascularization, initiation of mineral deposition, and combinatory acceleration of full-thickness osseous regeneration. This study demonstrated that Micromotion Biomaterials with controllable mechanophysical characteristics could promote the osteogenic differentiation of BMSCs and facilitate full osseous regeneration. The design of NIPAM/Nb2C hydrogel with highly efficient photothermal conversion, specific features of precisely controlled micromotion, and bionic-mimicking bone-repair capabilities could spark a new era in the field of regenerative medicine.
Collapse
Affiliation(s)
- Qianhao Yang
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Mengqiao Xu
- Eye Institute and Department of Ophthalmology, Eye and ENT Hospital, Fudan University, Shanghai, 200031, China
| | - Haoyu Fang
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Youshui Gao
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Daoyu Zhu
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Jing Wang
- Eye Institute and Department of Ophthalmology, Eye and ENT Hospital, Fudan University, Shanghai, 200031, China
| | - Yixuan Chen
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| |
Collapse
|
2
|
Liu W, Zou M, Chen M, Zhang Z, Mao Y, Yang Y, Liu Y, Shi Q, Wang X, Zhang F. Hypoxic environment promotes angiogenesis and bone bridge formation by activating Notch/RBPJ signaling pathway in HUVECs. Genomics 2024; 116:110838. [PMID: 38537807 DOI: 10.1016/j.ygeno.2024.110838] [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: 11/22/2023] [Revised: 02/09/2024] [Accepted: 03/24/2024] [Indexed: 05/27/2024]
Abstract
After epiphyseal fracture, the epiphyseal plate is prone to ischemia and hypoxia, leading to the formation of bone bridge and deformity. However, the exact mechanism controlling the bone bridge formation remains unclear. Notch/RBPJ signaling axis has been indicated to regulate angiogenesis and osteogenic differentiation. Our study aims to investigate the mechanism of bone bridge formation after epiphyseal plate injury, and to provide a theoretical basis for new therapeutic approaches to prevent the bone bridge formation. The expression of DLL4 and RBPJ was significantly up-regulated in HUVECs after ischemia and hypoxia treatment. Notch/RBPJ pathway positively regulated the osteogenic differentiation of BMSCs. HUVECs can induce osteogenic differentiation of BMSCs under ischemia and hypoxia. Notch/RBPJ pathway is involved in the regulation of the trans-epiphyseal bridge formation. Notch/RBPJ in HUVECs is associated with osteogenic differentiation of BMSCs and may participate in the regulation of the bone bridge formation across the epiphyseal plate.
Collapse
Affiliation(s)
- Wendong Liu
- Department of Orthopaedics, Children's Hospital of Soochow University, 92 Zhongnan St., Suzhou 215000, Jiangsu Province, China; Clinical Pediatrics School, Soochow University, 92 Zhongnan St., Suzhou 215000, Jiangsu Province, China
| | - Mincheng Zou
- Department of Orthopaedics, Children's Hospital of Soochow University, 92 Zhongnan St., Suzhou 215000, Jiangsu Province, China; Clinical Pediatrics School, Soochow University, 92 Zhongnan St., Suzhou 215000, Jiangsu Province, China
| | - Mimi Chen
- Department of Orthopaedics, Children's Hospital of Soochow University, 92 Zhongnan St., Suzhou 215000, Jiangsu Province, China
| | - Zheng Zhang
- Department of Orthopaedics, Children's Hospital of Soochow University, 92 Zhongnan St., Suzhou 215000, Jiangsu Province, China
| | - Yunpeng Mao
- Department of Orthopaedics, Children's Hospital of Soochow University, 92 Zhongnan St., Suzhou 215000, Jiangsu Province, China
| | - Yuhao Yang
- Department of Orthopaedics, Children's Hospital of Soochow University, 92 Zhongnan St., Suzhou 215000, Jiangsu Province, China
| | - Ya Liu
- Department of Orthopaedics, Children's Hospital of Soochow University, 92 Zhongnan St., Suzhou 215000, Jiangsu Province, China
| | - Qin Shi
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Soochow University, 188 Shizi St., Suzhou 215006, Jiangsu, China
| | - Xiaodong Wang
- Department of Orthopaedics, Children's Hospital of Soochow University, 92 Zhongnan St., Suzhou 215000, Jiangsu Province, China; Clinical Pediatrics School, Soochow University, 92 Zhongnan St., Suzhou 215000, Jiangsu Province, China
| | - Fuyong Zhang
- Department of Orthopaedics, Children's Hospital of Soochow University, 92 Zhongnan St., Suzhou 215000, Jiangsu Province, China.
| |
Collapse
|
3
|
Xu J, He S, Xia T, Shan Y, Wang L. Targeting type H vessels in bone-related diseases. J Cell Mol Med 2024; 28:e18123. [PMID: 38353470 PMCID: PMC10865918 DOI: 10.1111/jcmm.18123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 01/01/2024] [Accepted: 01/09/2024] [Indexed: 02/16/2024] Open
Abstract
Blood vessels are essential for bone development and metabolism. Type H vessels in bone, named after their high expression of CD31 and Endomucin (Emcn), have recently been reported to locate mainly in the metaphysis, exhibit different molecular properties and couple osteogenesis and angiogenesis. A strong correlation between type H vessels and bone metabolism is now well-recognized. The crosstalk between type H vessels and osteoprogenitor cells is also involved in bone metabolism-related diseases such as osteoporosis, osteoarthritis, fracture healing and bone defects. Targeting the type H vessel formation may become a new approach for managing a variety of bone diseases. This review highlighted the roles of type H vessels in bone-related diseases and summarized the research attempts to develop targeted intervention, which will help us gain a better understanding of their potential value in clinical application.
Collapse
Affiliation(s)
- Juan Xu
- Outpatient DepartmentChildren's Hospital of Soochow UniversitySuzhouChina
| | - Shuang‐jian He
- Department of OrthopaedicsSuzhou Hospital, Affiliated Hospital of Medical School, Nanjing UniversitySuzhouChina
| | - Ting‐ting Xia
- Clinical Research InstituteSuzhou Hospital, Affiliated Hospital of Medical School, Nanjing UniversitySuzhouChina
| | - Yu Shan
- Department of OrthopeadicsSuzhou Ninth Hospital Affiliated to Soochow UniversitySuzhouChina
| | - Liang Wang
- Department of OrthopaedicsSuzhou Hospital, Affiliated Hospital of Medical School, Nanjing UniversitySuzhouChina
- Department of OrthopeadicsThe Fourth Affiliated Hospital of Soochow UniversitySuzhouChina
| |
Collapse
|
4
|
Caliani Carrera AL, Minto BW, Malard P, Brunel HDSS. The Role of Mesenchymal Stem Cell Secretome (Extracellular Microvesicles and Exosomes) in Animals' Musculoskeletal and Neurologic-Related Disorders. Vet Med Int 2023; 2023:8819506. [PMID: 38023428 PMCID: PMC10645499 DOI: 10.1155/2023/8819506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 10/23/2023] [Accepted: 10/26/2023] [Indexed: 12/01/2023] Open
Abstract
The advances in regenerative medicine are very important for the development of medicine and the discovery of stem cells has shown a greater capacity to raise the level of therapeutic quality while their use becomes more accessible, especially in their mesenchymal form. In veterinary medicine, it is not different. The use of those cells, as well as recent advances related to the use of their extracellular vesicles, demonstrates a great opportunity to enhance therapeutic methods and ensure more life quality for patients, which can be in clinical or surgical treatments. Knowing the advances in these modalities and the growing clinical and surgery research and demands for innovations in orthopedic and neurology medicines, this paper aimed to review the literature about the methodologies of use and applications such as the pathways of action and the advances that were postulated for microvesicles and exosomes derived from mesenchymal stem cells in veterinary medicine, especially for musculoskeletal disorders and related injuries.
Collapse
Affiliation(s)
- Alefe Luiz Caliani Carrera
- Department of Clinical and Veterinary Surgery, São Paulo State University (UNESP), Av Paulo Donato Castelane s/n, Jaboticabal, São Paulo, Brazil
| | - Bruno Watanabe Minto
- Department of Clinical and Veterinary Surgery, São Paulo State University (UNESP), Av Paulo Donato Castelane s/n, Jaboticabal, São Paulo, Brazil
| | - Patrícia Malard
- Catholic University of Brasilia, Brasília, Federal District, Brazil
| | | |
Collapse
|
5
|
Chen Z, Zhao H, Meng L, Yu S, Liu Z, Xue J. Microfibril-Associated Glycoprotein-2 Promoted Fracture Healing via Integrin αvβ3/PTK2/AKT Signaling. J Transl Med 2023; 103:100121. [PMID: 36934797 DOI: 10.1016/j.labinv.2023.100121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 02/08/2023] [Accepted: 02/16/2023] [Indexed: 03/19/2023] Open
Abstract
Fracture healing is a complex physiological process in which angiogenesis plays an essential role. Microfibril-associated glycoprotein-2 (MAGP2) has been reported to possess a proangiogenic activity via integrin αvβ3, yet its role in bone repair is unexplored. In this study, a critical-sized femoral defect (2 mm) was created in mice, followed by the delivery of an adenovirus-based MAGP2 overexpression vector or its negative control at the fracture site. At days 7, 14, 21, and 28 postfracture, bone fracture healing was evaluated by radiography, micro-computed tomography, and histopathologic analysis. Adenovirus-based MAGP2 overexpression vector-treated mice exhibited increased bone mineral density and bone volume fraction. MAGP2 overexpression contributed to an advanced stage of endochondral ossification and induced cartilage callus into the bony callus. Further analysis indicated that MAGP2 was associated with enhanced angiogenesis, as evidenced by marked MAGP2 and integrin αvβ3 costaining and increased endothelial cell markers such as endomucin and CD31 levls, as well as elevated phosphorylation of protein tyrosine kinase 2 (PTK2) and AKT serine/threonine kinase 1 (AKT) in the callus. In vitro, recombinant human MAGP2 treatment enhanced the viability, migration, and tube formation ability of human microvascular endothelial cells, which was partially reversed by integrin αvβ3 inhibition or MK-2206, a specific AKT inhibitor. Inhibition of integrin αvβ3 abolished MAGP2-induced PTK2 and AKT activation. Taken together, our data provide the first evidence that MAGP2 promotes angiogenesis and bone formation by activating the integrin αvβ3/PTK2/AKT signaling pathway.
Collapse
Affiliation(s)
- Zhiguang Chen
- Department of Emergency Medicine, Shengjing Hospital of China Medical University, Shenyang, China
| | - Haibin Zhao
- Department of Emergency Medicine, Shengjing Hospital of China Medical University, Shenyang, China
| | - Lingshuai Meng
- Department of Emergency Medicine, Shengjing Hospital of China Medical University, Shenyang, China
| | - Shengwei Yu
- Department of Emergency Medicine, Shengjing Hospital of China Medical University, Shenyang, China
| | - Zhenning Liu
- Department of Emergency Medicine, Shengjing Hospital of China Medical University, Shenyang, China
| | - Jinqi Xue
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
6
|
Migliorini F, Cocconi F, Vecchio G, Schäefer L, Koettnitz J, Maffulli N. Pharmacological agents for bone fracture healing: talking points from recent clinical trials. Expert Opin Investig Drugs 2023; 32:855-865. [PMID: 37740660 DOI: 10.1080/13543784.2023.2263352] [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: 04/18/2023] [Accepted: 09/22/2023] [Indexed: 09/24/2023]
Abstract
INTRODUCTION Pharmacological strategies might influence bone healing in terms of time to union or quality of mature bone. This expert opinion discussed the current level I evidence on the experimental pharmacological agents used to favor bone fracture healing. AREAS COVERED This study was conducted according to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses: the 2020 PRISMA statement. In April 2023, the following databases were accessed: PubMed, Web of Science, Google Scholar, Embase. All the randomized clinical trials investigating pharmacological agents for bone fracture healing were accessed. No time constraint was set for the search. The search was restricted to RCTs. No additional filters were used in the database search. Data from 19 RCTs (4067 patients) were collected. 78% (3160 of 4067) were women. The mean length of the follow-up was 9.3 months (range, 1-26 months). The mean age of the patients was 64.4 years (range, 8-84 years). EXPERT OPINION Calcitonin could favor bone fracture healing. Bisphosphonates (alendronate, zoledronate, clodronate), monoclonal antibodies (denosumab, romosozumab), statins, vitamin D and calcium supplementation, strontium ranelate, and ibuprofen did not influence bony healing. Concerning the effect of parathormone, current level I evidence is controversial, and additional studies are required. LEVEL OF EVIDENCE Level I, systematic review of RCTs.
Collapse
Affiliation(s)
- Filippo Migliorini
- Department of Orthopaedic, Trauma, and Reconstructive Surgery, RWTH University Medical Centre, Aachen, Germany
| | - Federico Cocconi
- Department of Orthopedics and Trauma Surgery, Academic Hospital of Bolzano (SABES-ASDAA), Teaching Hospital of Paracelsus Medical University, Bolzano, Italy
| | - Gianluca Vecchio
- Department of Trauma and Orthopaedic Surgery, University Hospital Sant' Andrea, University La Sapienza, Rome, Italy
| | - Luise Schäefer
- Department of Orthopaedic, Trauma, and Reconstructive Surgery, RWTH University Medical Centre, Aachen, Germany
| | - Julian Koettnitz
- Department of Orthopedics, Auguste-Viktoria Clinic, Ruhr University Bochum, Bad Oeynhausen, Germany
| | - Nicola Maffulli
- Department of Trauma and Orthopaedic Surgery, University Hospital Sant' Andrea, University La Sapienza, Rome, Italy
- School of Pharmacy and Bioengineering, Keele University Faculty of Medicine, Stoke on Trent, UK
- Centre for Sports and Exercise Medicine, Barts and the London School of Medicine and Dentistry, Mile End Hospital, Queen Mary University of London, London, UK
| |
Collapse
|
7
|
Hu J, Shao J, Huang G, Zhang J, Pan S. In Vitro and In Vivo Applications of Magnesium-Enriched Biomaterials for Vascularized Osteogenesis in Bone Tissue Engineering: A Review of Literature. J Funct Biomater 2023; 14:326. [PMID: 37367290 DOI: 10.3390/jfb14060326] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 06/09/2023] [Accepted: 06/12/2023] [Indexed: 06/28/2023] Open
Abstract
Bone is a highly vascularized tissue, and the ability of magnesium (Mg) to promote osteogenesis and angiogenesis has been widely studied. The aim of bone tissue engineering is to repair bone tissue defects and restore its normal function. Various Mg-enriched materials that can promote angiogenesis and osteogenesis have been made. Here, we introduce several types of orthopedic clinical uses of Mg; recent advances in the study of metal materials releasing Mg ions (pure Mg, Mg alloy, coated Mg, Mg-rich composite, ceramic, and hydrogel) are reviewed. Most studies suggest that Mg can enhance vascularized osteogenesis in bone defect areas. Additionally, we summarized some research on the mechanisms related to vascularized osteogenesis. In addition, the experimental strategies for the research of Mg-enriched materials in the future are put forward, in which clarifying the specific mechanism of promoting angiogenesis is the crux.
Collapse
Affiliation(s)
- Jie Hu
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Jiahui Shao
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Gan Huang
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Jieyuan Zhang
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Shuting Pan
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| |
Collapse
|
8
|
Priddy LB, Krishnan L, Hettiaratchi MH, Karthikeyakannan S, Gupte N, Guldberg RE. Amniotic membrane attenuates heterotopic ossification following high-dose bone morphogenetic protein-2 treatment of segmental bone defects. J Orthop Res 2023; 41:130-140. [PMID: 35340049 PMCID: PMC9512937 DOI: 10.1002/jor.25324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 01/31/2022] [Accepted: 03/11/2022] [Indexed: 02/04/2023]
Abstract
Treatment of large bone defects with supraphysiological doses of bone morphogenetic protein-2 (BMP-2) has been associated with complications including heterotopic ossification (HO), inflammation, and pain, presumably due to poor spatiotemporal control of BMP-2. We have previously recapitulated extensive HO in our rat femoral segmental defect model by treatment with high-dose BMP-2 (30 μg). Using this model and BMP-2 dose, our objective was to evaluate the utility of a clinically available human amniotic membrane (AM) around the defect space for guided bone regeneration and reduction of HO. We hypothesized that AM surrounding collagen sponge would attenuate heterotopic ossification compared with collagen sponge alone. In vitro, AM retained more BMP-2 than a synthetic poly(ε-caprolactone) membrane through 21 days. In vivo, as hypothesized, the collagen + AM resulted in significantly less heterotopic ossification and correspondingly, lower total bone volume (BV), compared with collagen sponge alone. Although bone formation within the defect was delayed with AM around the defect, by 12 weeks, defect BVs were equivalent. Torsional stiffness was significantly reduced with AM but was equivalent to that of intact bone. Collagen + AM resulted in the formation of dense fibrous tissue and mineralized tissue, while the collagen group contained primarily mineralized tissue surrounded by marrow-like structures. Especially in conjunction with high doses of growth factor delivered via collagen sponge, these findings suggest AM may be effective as an overlay adjacent to bone healing sites to spatially direct bone regeneration and minimize heterotopic ossification.
Collapse
Affiliation(s)
- Lauren B. Priddy
- Parker H. Petit Institute for Bioengineering & Bioscience, Georgia Institute of Technology, 315 Ferst Drive NW, Atlanta, GA 30332, USA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, 313 Ferst Drive NW, Atlanta, GA 30332, USA
- Current affiliation: Department of Agricultural and Biological Engineering, Mississippi State University, 130 Creelman Street, Mississippi State, MS 39762, USA
| | - Laxminarayanan Krishnan
- Parker H. Petit Institute for Bioengineering & Bioscience, Georgia Institute of Technology, 315 Ferst Drive NW, Atlanta, GA 30332, USA
| | - Marian H. Hettiaratchi
- Parker H. Petit Institute for Bioengineering & Bioscience, Georgia Institute of Technology, 315 Ferst Drive NW, Atlanta, GA 30332, USA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, 313 Ferst Drive NW, Atlanta, GA 30332, USA
- Current affiliation: Phil and Penny Knight Campus for Accelerating Scientific Impact, University of Oregon, 6231 University of Oregon, Eugene, OR 97403, USA
| | - Sukhita Karthikeyakannan
- Parker H. Petit Institute for Bioengineering & Bioscience, Georgia Institute of Technology, 315 Ferst Drive NW, Atlanta, GA 30332, USA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, 313 Ferst Drive NW, Atlanta, GA 30332, USA
| | - Nikhil Gupte
- Parker H. Petit Institute for Bioengineering & Bioscience, Georgia Institute of Technology, 315 Ferst Drive NW, Atlanta, GA 30332, USA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, 313 Ferst Drive NW, Atlanta, GA 30332, USA
| | - Robert E. Guldberg
- Parker H. Petit Institute for Bioengineering & Bioscience, Georgia Institute of Technology, 315 Ferst Drive NW, Atlanta, GA 30332, USA
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, 801 Ferst Drive NW, Atlanta, GA 30332, USA
- Current affiliation: Phil and Penny Knight Campus for Accelerating Scientific Impact, University of Oregon, 6231 University of Oregon, Eugene, OR 97403, USA
| |
Collapse
|
9
|
Breulmann FL, Hatt LP, Schmitz B, Wehrle E, Richards RG, Della Bella E, Stoddart MJ. Prognostic and therapeutic potential of microRNAs for fracture healing processes and non-union fractures: A systematic review. Clin Transl Med 2023; 13:e1161. [PMID: 36629031 PMCID: PMC9832434 DOI: 10.1002/ctm2.1161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 12/13/2022] [Accepted: 12/19/2022] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Approximately 10% of all bone fractures result in delayed fracture healing or non-union; thus, the identification of biomarkers and prognostic factors is of great clinical interest. MicroRNAs (miRNAs) are known to be involved in the regulation of the bone healing process and may serve as functional markers for fracture healing. AIMS AND METHODS This systematic review aimed to identify common miRNAs involved in fracture healing or non-union fractures using a qualitative approach. A systematic literature search was performed with the keywords 'miRNA and fracture healing' and 'miRNA and non-union fracture'. Any original article investigating miRNAs in fracture healing or non-union fractures was screened. Eventually, 82 studies were included in the qualitative analysis for 'miRNA and fracture healing', while 19 were selected for the 'miRNA and fracture non-union' category. RESULTS AND CONCLUSIONS Out of 151 miRNAs, miR-21, miR-140 and miR-214 were the most investigated miRNAs in fracture healing in general. miR-31-5p, miR-221 and miR-451-5p were identified to be regulated specifically in non-union fractures. Large heterogeneity was detected between studies investigating the role of miRNAs in fracture healing or non-union in terms of patient population, sample types and models used. Nonetheless, our approach identified some miRNAs with the potential to serve as biomarkers for non-union fractures, including miR-31-5p, miR-221 and miR-451-5p. We provide a discussion of involved pathways and suggest on alignment of future research in the field.
Collapse
Affiliation(s)
- Franziska Lioba Breulmann
- AO Research Institute DavosDavos PlatzSwitzerland
- Department of Orthopedic Sports MedicineKlinikum Rechts der IsarTechnical University of MunichMunichGermany
| | - Luan Phelipe Hatt
- AO Research Institute DavosDavos PlatzSwitzerland
- Institute for BiomechanicsETH ZürichZurichSwitzerland
| | - Boris Schmitz
- Department of Rehabilitation SciencesFaculty of HealthUniversity of Witten/HerdeckeWittenGermany
- DRV Clinic KönigsfeldCenter for Medical RehabilitationEnnepetalGermany
| | - Esther Wehrle
- AO Research Institute DavosDavos PlatzSwitzerland
- Institute for BiomechanicsETH ZürichZurichSwitzerland
| | - Robert Geoff Richards
- AO Research Institute DavosDavos PlatzSwitzerland
- Faculty of MedicineMedical Center‐Albert‐Ludwigs‐University of FreiburgAlbert‐Ludwigs‐University of FreiburgFreiburgGermany
| | | | - Martin James Stoddart
- AO Research Institute DavosDavos PlatzSwitzerland
- Faculty of MedicineMedical Center‐Albert‐Ludwigs‐University of FreiburgAlbert‐Ludwigs‐University of FreiburgFreiburgGermany
| |
Collapse
|
10
|
Qin Q, Lee S, Patel N, Walden K, Gomez-Salazar M, Levi B, James AW. Neurovascular coupling in bone regeneration. Exp Mol Med 2022; 54:1844-1849. [PMID: 36446849 PMCID: PMC9722927 DOI: 10.1038/s12276-022-00899-6] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Revised: 10/11/2022] [Accepted: 10/11/2022] [Indexed: 11/30/2022] Open
Abstract
The mammalian skeletal system is densely innervated by both neural and vascular networks. Peripheral nerves in the skeleton include sensory and sympathetic nerves. The crosstalk between skeletal and neural tissues is critical for skeletal development and regeneration. The cellular processes of osteogenesis and angiogenesis are coupled in both physiological and pathophysiological contexts. The cellular and molecular regulation of osteogenesis and angiogenesis have yet to be fully defined. This review will provide a detailed characterization of the regulatory role of nerves and blood vessels during bone regeneration. Furthermore, given the importance of the spatial relationship between nerves and blood vessels in bone, we discuss neurovascular coupling during physiological and pathological bone formation. A better understanding of the interactions between nerves and blood vessels will inform future novel therapeutic neural and vascular targeting for clinical bone repair and regeneration.
Collapse
Affiliation(s)
- Qizhi Qin
- grid.21107.350000 0001 2171 9311Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205 USA
| | - Seungyong Lee
- grid.260024.20000 0004 0627 4571Department of Physiology, College of Graduate Studies, Midwestern University, Glendale, AZ 85308 USA ,grid.412977.e0000 0004 0532 7395Department of Physical Education, Incheon National University, Incheon, 22012 South Korea
| | - Nirali Patel
- grid.260024.20000 0004 0627 4571Arizona College of Osteopathic Medicine, Midwestern University, Glendale, AZ 85308 USA
| | - Kalah Walden
- grid.260024.20000 0004 0627 4571Arizona College of Osteopathic Medicine, Midwestern University, Glendale, AZ 85308 USA
| | - Mario Gomez-Salazar
- grid.21107.350000 0001 2171 9311Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205 USA
| | - Benjamin Levi
- grid.267313.20000 0000 9482 7121Departments of Surgery, UT Southwestern Medical Center, Dallas, TX 75390 USA
| | - Aaron W. James
- grid.21107.350000 0001 2171 9311Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205 USA
| |
Collapse
|
11
|
Liu L, Cai B, Liu L, Zhuang X, Wu Y, Zhang J. The effect of soft tissue defect on callus formation in Kunming mice different tibial injury models.. [DOI: 10.21203/rs.3.rs-2006802/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Abstract
Objective: To investigated the highly replicable bone injury model and the effect of soft tissue defect on bone repair. Methods: Fifty 6-week-old female kunming mice were randomly divided into 5 groups, and the 4 groups of them underwent fracture or bone defect surgery on the right tibia with or without tibialis anterior muscle defect respectively. The right injured tibias and heart blood were collected on day 10 after operation for Micro-CT, histological and ELISA analysis respectively. The fifth group was control group,and the cardiac blood was collected at the same time for ELISA.RESULTS: Micro-CT and histological examination indicated that our modelling approach could successfully provide different animal models of open bone injury. Micro-CT showed a significant increase in the ratio of bone volume to tissue volume (BV/TV, %) after soft tissue defect in different bone injury models compared to the soft tissue preserved group. Histomorphometric analysis demonstrated a significant increase in the amount of total bone callus, cartilage callus and fibrous tissue after soft tissue defects, while the amount of hard callus was significantly reduced. Immunohistochemical analysis showed higher levels of CYR61 and VEGFR2 after soft tissue defect. ELISA results revealed no significant difference in IL-1β levels between the soft tissue preserved and soft tissue defect groups. HE staining also confirmed no significant difference in the degree of inflammatory cell infiltration after soft tissue defect.Conclusion: The above models were simple,highly reproducible, and provided reliable animal models for studying the bone healing. We inferred that mechanical stability played an important role in the process of bone healing, and the soft tissue around the injury site mainly provided fixation and protection.
Collapse
Affiliation(s)
- Lingling Liu
- The First Affiliated Hospital of Shantou University Medical College
| | - Bozhi Cai
- The First Affiliated Hospital of Shantou University Medical College
| | - Liping Liu
- The First Affiliated Hospital of Shantou University Medical College
| | - Xiaoning Zhuang
- The First Affiliated Hospital of Shantou University Medical College
| | - Yanlan Wu
- The First Affiliated Hospital of Shantou University Medical College
| | - Jianfa Zhang
- The First Affiliated Hospital of Shantou University Medical College
| |
Collapse
|
12
|
Dou Y, Fang Y, Zhao C, Fu W, Jiang D. Editorial: Bioengineering and translational research for bone and joint diseases. Front Bioeng Biotechnol 2022; 10:969416. [PMID: 36091436 PMCID: PMC9459223 DOI: 10.3389/fbioe.2022.969416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 06/27/2022] [Indexed: 01/09/2023] Open
Affiliation(s)
- Yun Dou
- Peking University Third Hospital, Beijing, China
| | - Yin Fang
- Nanyang Technological University, Singapore, Singapore
| | - Chao Zhao
- University of Alabama, Tuscaloosa, AL, United States
| | - Weili Fu
- West China Hospital, Sichuan University, Chengdu, China
| | - Dong Jiang
- Peking University Third Hospital, Beijing, China,*Correspondence: Dong Jiang,
| |
Collapse
|
13
|
Zhang S, Tuk B, van de Peppel J, Kremers GJ, Koedam M, Pesch GR, Rahman Z, Hoogenboezem RM, Bindels EMJ, van Neck JW, Boukany PE, van Leeuwen JPTM, van der Eerden BCJ. Microfluidic evidence of synergistic effects between mesenchymal stromal cell-derived biochemical factors and biomechanical forces to control endothelial cell function. Acta Biomater 2022; 151:346-359. [PMID: 35995408 DOI: 10.1016/j.actbio.2022.08.025] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 07/20/2022] [Accepted: 08/12/2022] [Indexed: 11/01/2022]
Abstract
A functional vascular system is a prerequisite for bone repair as disturbed angiogenesis often causes non-union. Paracrine factors released from human bone marrow derived mesenchymal stromal cells (BMSCs) have angiogenic effects on endothelial cells. However, whether these paracrine factors participate in blood flow dynamics within bone capillaries remains poorly understood. Here, we used two different microfluidic designs to investigate critical steps during angiogenesis and found pronounced effects of endothelial cell proliferation as well as chemotactic and mechanotactic migration induced by BMSC conditioned medium (CM). The application of BMSC-CM in dynamic cultures demonstrates that bioactive factors in combination with fluidic flow-induced biomechanical signals significantly enhanced endothelial cell migration. Transcriptional analyses of endothelial cells demonstrate the induction of a unique gene expression profile related to tricarboxylic acid cycle and energy metabolism by the combination of BMSC-CM factors and shear stress, which opens an interesting avenue to explore during fracture healing. Our results stress the importance of in vivo - like microenvironments simultaneously including biochemical, biomechanical and oxygen levels when investigating key events during vessel repair. STATEMENT OF SIGNIFICANCE: Our results demonstrate the importance of recapitulating in vivo - like microenvironments when investigating key events during vessel repair. Endothelial cells exhibit enhanced angiogenesis characteristics when simultaneous exposing them to hMSC-CM, mechanical forces and biochemical signals simultaneously. The improved angiogenesis may not only result from the direct effect of growth factors, but also by reprogramming of endothelial cell metabolism. Moreover, with this model we demonstrated a synergistic impact of mechanical forces and biochemical factors on endothelial cell behavior and the expression of genes involved in the TCA cycle and energy metabolism, which opens an interesting new avenue to stimulate angiogenesis during fracture healing.
Collapse
Affiliation(s)
- Shuang Zhang
- Laboratory for Calcium and Bone Metabolism, Department of Internal Medicine, Erasmus University Medical Center; Rotterdam, the Netherlands
| | - Bastiaan Tuk
- Department of Plastic and Reconstructive Surgery, Erasmus University Medical Center; Rotterdam, the Netherlands
| | - Jeroen van de Peppel
- Laboratory for Calcium and Bone Metabolism, Department of Internal Medicine, Erasmus University Medical Center; Rotterdam, the Netherlands
| | - Gert-Jan Kremers
- Erasmus Optical Imaging Center, Erasmus University Medical Center; Rotterdam, the Netherlands
| | - Marijke Koedam
- Laboratory for Calcium and Bone Metabolism, Department of Internal Medicine, Erasmus University Medical Center; Rotterdam, the Netherlands
| | - Georg R Pesch
- Department of Chemical Engineering, Delft University of Technology; Delft, the Netherlands
| | - Zaid Rahman
- Department of Chemical Engineering, Delft University of Technology; Delft, the Netherlands
| | - Remco M Hoogenboezem
- Department of Hematology, Erasmus University Medical Center; Rotterdam, the Netherlands
| | - Eric M J Bindels
- Department of Hematology, Erasmus University Medical Center; Rotterdam, the Netherlands
| | - Johan W van Neck
- Department of Plastic and Reconstructive Surgery, Erasmus University Medical Center; Rotterdam, the Netherlands
| | - Pouyan E Boukany
- Department of Chemical Engineering, Delft University of Technology; Delft, the Netherlands
| | - Johannes P T M van Leeuwen
- Laboratory for Calcium and Bone Metabolism, Department of Internal Medicine, Erasmus University Medical Center; Rotterdam, the Netherlands
| | - Bram C J van der Eerden
- Laboratory for Calcium and Bone Metabolism, Department of Internal Medicine, Erasmus University Medical Center; Rotterdam, the Netherlands.
| |
Collapse
|
14
|
Liu M, Shafiq M, Sun B, Wu J, Wang W, El-Newehy M, El-Hamshary H, Morsi Y, Ali O, Khan AUR, Mo X. Composite Superelastic Aerogel Scaffolds Containing Flexible SiO 2 Nanofibers Promote Bone Regeneration. Adv Healthc Mater 2022; 11:e2200499. [PMID: 35670086 DOI: 10.1002/adhm.202200499] [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: 03/04/2022] [Revised: 04/25/2022] [Indexed: 11/08/2022]
Abstract
Repairing irregular-shaped bone defects poses enormous challenges. Scaffolds that can fully fit the defect site and simultaneously induce osteogenesis and angiogenesis hold great promise for bone defect healing. This study aimed to produce superelastic organic/inorganic composite aerogel scaffolds by blending silica nanofibers (SiO2 ) and poly (lactic acid)/gelatin (PLA/gel) nanofibers; the content of SiO2 nanofibers were varied from 0-60 wt% (e.g., PLA/gel, PLA/gel/SiO2 -L, PLA/gel/SiO2 -M, and PLA/gel/SiO2 -H for 0%, 20%, 40%, and 60% of SiO2 nanofibers, respectively) to produce a range of scaffolds. The PLA/gel/SiO2 -M scaffold had excellent elasticity and good mechanical properties. In vitro experiments demonstrated that the silicon ions released from PLA/gel/SiO2 -M scaffolds could promote the differentiation of rat bone marrow-derived mesenchymal stem cells (rBMSCs) into osteoblasts, thereby enhancing alkaline phosphatase activity and bone-related genes expressions. Meanwhile, the released silicon ions also promoted the proliferation of human umbilical vein endothelial cells (HUVECs) and the expression of vascular endothelial growth factors, thereby promoting angiogenesis. The assessment of these scaffolds in a calvarial defect model in rats showed good potential of PLA/gel/SiO2 -M to induce bone regeneration as well as promote osteogenesis and angiogenesis. Overall, these superelastic scaffolds containing flexible SiO2 nanofibers can simultaneously induce osteogenesis and angiogenesis, which may have broad applications for tissue engineering applications. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Mingyue Liu
- Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai, 201620, P. R. China
| | - Muhammad Shafiq
- Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai, 201620, P. R. China
| | - Binbin Sun
- Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai, 201620, P. R. China
| | - Jinglei Wu
- Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai, 201620, P. R. China
| | - Wei Wang
- Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai, 201620, P. R. China
| | - Mohamed El-Newehy
- Department of Chemistry, College of Science, King Saud University, P.O. Box 2455, Riyadh, 11451, Saudi Arabia
| | - Hany El-Hamshary
- Department of Chemistry, College of Science, King Saud University, P.O. Box 2455, Riyadh, 11451, Saudi Arabia
| | - Yosry Morsi
- Faculty of Engineering and Industrial Sciences, Swinburne University of Technology, Boroondara, VIC, 3122, Australia
| | - Onaza Ali
- School of Chemistry and Chemical Engineering, Tiangong University, Tianjin, 300387, China
| | - Atta Ur Rehman Khan
- Department of Biotechnology, The University of Azad Jammu & Kashmir, Muzaffarabad, Pakistan
| | - Xiumei Mo
- Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai, 201620, P. R. China
| |
Collapse
|
15
|
Hang R, Tian X, Qu G, Zhao Y, Yao R, Zhang Y, Wei W, Yao X, Chu PK. Exosomes derived from magnesium ion-stimulated macrophages inhibit angiogenesis. Biomed Mater 2022; 17. [PMID: 35477160 DOI: 10.1088/1748-605x/ac6b03] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Accepted: 04/27/2022] [Indexed: 11/11/2022]
Abstract
Angiogenesis, an essential prerequisite to osteogenesis in bone repair and regeneration, can be mediated by immunoregulation of macrophages. Magnesium and its alloys are promising biodegradable bone implant materials and can affect immunoregulation of macrophages by the degradation products (magnesium ions. Nevertheless, the mechanism of macrophage-derived exosomes stimulated by Mg ions in immunoregulation is still not well understood. Herein, 10-50 mM magnesium ions are shown to inhibit the macrophage viability and proliferation in a dose-dependent manner, but a high concentration results in macrophage apoptosis. The exosomes secreted by macrophages from magnesium ion stimulation inhibit angiogenesis of endothelial cells, as manifested by the suppressed cell viability, proliferation, migration, and tube formation, which arise at least partially from exosome-mediated downregulation of endothelial nitric oxide and the vascular endothelial growth factor. The findings reported in this paper suggest that the bio-functionality of biodegradable magnesium alloys must be considered from the perspective of immunoregulation of macrophage-derived exosomes. Our results also suggest potential cancer therapy by inhibiting tumor-associated angiogenesis.
Collapse
Affiliation(s)
- Ruiqiang Hang
- Taiyuan University of Technology, No. 79, Yingze West Road, Taiyuan, Shanxi Province, Taiyuan, 030024, CHINA
| | - Xue Tian
- Taiyuan University of Technology, No. 79, Yingze West Road, Taiyuan, Shanxi Province, Taiyuan, 030024, CHINA
| | - Guangping Qu
- Taiyuan University of Technology, No. 79, Yingze West Road, Taiyuan, Shanxi Province, Taiyuan, 030024, CHINA
| | - Yuyu Zhao
- Taiyuan University of Technology, No. 79, Yingze West Road, Taiyuan, Shanxi Province, Taiyuan, 030024, CHINA
| | - Runhua Yao
- Taiyuan University of Technology, No. 79, Yingze West Road, Taiyuan, Shanxi Province, Taiyuan, 030024, CHINA
| | - Yi Zhang
- Taiyuan University of Technology, No. 79, Yingze West Road, Taiyuan, Shanxi Province, Taiyuan, 030024, CHINA
| | - Wenfa Wei
- Taiyuan University of Technology, No. 79, Yingze West Road, Taiyuan, Shanxi Province, Taiyuan, 030024, CHINA
| | - Xiaohong Yao
- Taiyuan University of Technology, No. 79, Yingze West Road, Taiyuan, Shanxi Province, Taiyuan, 030024, CHINA
| | - Paul K Chu
- Department of Physics and Materials Science, City University of Hong Kong, Tat Chee Avenue, Kowloon, HONG KONG
| |
Collapse
|
16
|
Ding L, Gu S, Zhou B, Wang M, Zhang Y, Wu S, Zou H, Zhao G, Gao Z, Xu L. Ginsenoside Compound K Enhances Fracture Healing via Promoting Osteogenesis and Angiogenesis. Front Pharmacol 2022; 13:855393. [PMID: 35462912 PMCID: PMC9020191 DOI: 10.3389/fphar.2022.855393] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Accepted: 03/07/2022] [Indexed: 11/13/2022] Open
Abstract
Fractures have an extraordinarily negative impact on an individual’s quality of life and functional status, particularly delayed or non-union fractures. Osteogenesis and angiogenesis are closely related to bone growth and regeneration, and bone modeling and remodeling. Recently Chinese medicine has been extensively studied to promote osteogenic differentiation in MSCs. Studies have found that Ginseng can be used as an alternative for tissue regeneration and engineering. Ginseng is a commonly used herbal medicine in clinical practice, and one of its components, Ginsenoside Compound K (CK), has received much attention. Evidence indicates that CK has health-promoting effects in inflammation, atherosclerosis, diabetics, aging, etc. But relatively little is known about its effect on bone regeneration and the underlying cellular and molecular mechanisms. In this study, CK was found to promote osteogenic differentiation of rat bone marrow mesenchymal stem cells (rBMSCs) by RT-PCR and Alizarin Red S staining in vitro. Mechanistically, we found CK could promote osteogenesis through activating Wnt/β-catenin signaling pathway by immunofluorescence staining and luciferase reporter assay. And we also showed that the tube formation capacity of human umbilical vein endothelial cells (HUVECs) was increased by CK. Furthermore, using the rat open femoral fracture model, we found that CK could improve fracture repair as demonstrated by Micro-CT, biomechanical and histology staining analysis. The formation of H type vessel in the fracture callus was also increased by CK. These findings provide a scientific basis for treating fractures with CK, which may expand its application in clinical practice.
Collapse
Affiliation(s)
- Lingli Ding
- Key Laboratory of Orthopaedics and Traumatology, Lingnan Medical Research Center, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Song Gu
- The First Affiliated Hospital, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Bingyu Zhou
- Key Laboratory of Orthopaedics and Traumatology, Lingnan Medical Research Center, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Min Wang
- Key Laboratory of Orthopaedics and Traumatology, Lingnan Medical Research Center, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yage Zhang
- Key Laboratory of Orthopaedics and Traumatology, Lingnan Medical Research Center, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Siluo Wu
- Key Laboratory of Orthopaedics and Traumatology, Lingnan Medical Research Center, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Hong Zou
- Engineering Laboratory for Nutrition, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, China
| | - Guoping Zhao
- Master Lab for Innovative Application of Nature Products, National Center of Technology Innovation for Synthetic Biology, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, China
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- CAS-Key Laboratory of Synthetic Biology, CAS Center for Excellence in Molecular Plant Sciences, Shanghai Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai, China
- Bio-Med Big Data Center, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, China
- State Key Laboratory of Genetic Engineering, Department of Microbiology and Immunology, School of Life Sciences, Fudan University, Shanghai, China
- Department of Microbiology, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
- *Correspondence: Guoping Zhao, ; Zhao Gao, ; Liangliang Xu,
| | - Zhao Gao
- Er Sha Sports Training Center of Guangdong Province, Guangzhou, China
- *Correspondence: Guoping Zhao, ; Zhao Gao, ; Liangliang Xu,
| | - Liangliang Xu
- Key Laboratory of Orthopaedics and Traumatology, Lingnan Medical Research Center, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
- *Correspondence: Guoping Zhao, ; Zhao Gao, ; Liangliang Xu,
| |
Collapse
|
17
|
Chen C, Yan S, Geng Z, Wang Z. Fracture repair by IOX2: Regulation of the hypoxia inducible factor-1α signaling pathway and BMSCs. Eur J Pharmacol 2022; 921:174864. [PMID: 35219731 DOI: 10.1016/j.ejphar.2022.174864] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Accepted: 02/22/2022] [Indexed: 11/03/2022]
Abstract
The treatment of fracture delayed union and nonunion has become a challenging problem. Hypoxia inducible factor-1α (HIF-1α) is reported to be a key factor in fracture healing, and is degraded by hydroxylation of prolyl hydroxylase (PHDs) under normal oxygen. Small molecules could inhibit the activity of PHDs, stabilize HIF-1α protein, regulate the expression of downstream target genes of HIF-1α, and make the body adapt to hypoxia. The migration and osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs) is the most promising candidate for the treatment of fracture nonunion. Here we reported that IOX2, an HIF-1α PHD inhibitor, markedly improved the proliferation and migration of BMSCs by upregulating intracellular Ca2+ and concomitant decreasing reactive oxygen species (ROS) in vitro, and facilitated the repair of bone fracture by increasing the number of BMSCs and cartilage formation in vivo. No significant influence of IOX2 on the proliferation and migration of BMSCs after silencing of the HIF-1α. Together, our findings indicated that IOX2 promoted the proliferation and migration of BMSCs via the HIF-1α pathway and further accelerated fracture healing. These results provide a deeper understanding of the mechanism by which HIF promotes fracture healing.
Collapse
Affiliation(s)
- Chunxia Chen
- State Key Laboratory of Coordination Chemistry, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China; Joint National Laboratory for Antibody Drug Engineering, The First Affiliated Hospital, Henan University, Kaifeng, 475004, China
| | - Shihai Yan
- State Key Laboratory of Coordination Chemistry, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China; Department of Pharmacology, Jiangsu Province Hospital of Chinese Medicine, Nanjing, 210029, China
| | - Zhirong Geng
- State Key Laboratory of Coordination Chemistry, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Zhilin Wang
- State Key Laboratory of Coordination Chemistry, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China.
| |
Collapse
|
18
|
Zhou Z, Yan Y, Yu H, Yang G, Su H, Zhang T, Fan Y, Zhao F. Effect of Inter-Fragmentary Gap Size on Neovascularization During Bone Healing: A Micro-CT Imaging Study. Front Bioeng Biotechnol 2022; 10:808182. [PMID: 35345467 PMCID: PMC8957065 DOI: 10.3389/fbioe.2022.808182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 01/19/2022] [Indexed: 12/03/2022] Open
Abstract
Introduction: Neovascularization of the fracture site is of great importance for bone healing and could be influenced by local mechanical environment such as fixation stability and inter-fragmentary gap size. This study aims to reconstruct the neovascularization of the fracture site and explore the effect of inter-fragmentary gap size on the spatiotemporal structure of vascularity during bone healing. Methods: Osteotomy was performed on 36 Sprague–Dawley (SD) rats on the right tibial diaphysis, and the fracture was given stable fixation with two different inter-fragmentary gap sizes. SD rats received stable fixation with either a small-sized inter-fragmentary gap (FSF1, 1 mm, n = 18) or a large-sized one (FSF3, 3 mm, n = 18). The left hind limbs were treated as the control group (CON). The animals were killed at different time points (2, 4, and 6 weeks postoperatively, n = 6, respectively) for vascular perfusion and micro-CT imaging. Results: (a) At week 2 and 4, FSF1 group showed significantly higher vessel volume ratio (VV/TV) and vessel surface density (VS/TV) values than both CON and FSF3 group; there was no significant difference in either VV/TV or VS/TV values between CON and FSF3 groups. (b) At week 6, both FSF1 and FSF3 groups showed significantly higher VV/TV and VS/TV values than CON group; FSF3 group had a significantly higher VV/TV value than FSF1 group. Conclusion: Different inter-fragmentary gap sizes greatly affect the timing of angiogenesis at the fracture site. Stable fixation with a small inter-fragmentary gap (1 mm) benefits neovascularization at the early stages during bone healing and reconstruction, while stable fixation with a large inter-fragmentary gap (3 mm) delays the occurrence of angiogenesis to a later phase.
Collapse
Affiliation(s)
- Zhilun Zhou
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Yang Yan
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Hao Yu
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Guanzhong Yang
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Hao Su
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Tao Zhang
- Department of Orthopeadics, Tianjin Hospital, Tianjin, China
- *Correspondence: Tao Zhang, ; Yubo Fan, ; Feng Zhao,
| | - Yubo Fan
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
- *Correspondence: Tao Zhang, ; Yubo Fan, ; Feng Zhao,
| | - Feng Zhao
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
- *Correspondence: Tao Zhang, ; Yubo Fan, ; Feng Zhao,
| |
Collapse
|
19
|
Abstract
Despite major research efforts to elucidate mechanisms of non-union formation, failed fracture healing remains a common complication in orthopedic surgery. Adequate vascularization has been recognized as a crucial factor for successful bone regeneration, as newly formed microvessels guarantee the supply of the callus tissue with vital oxygen, nutrients, and growth factors. Accordingly, a vast number of preclinical studies have focused on the development of vascularization strategies to stimulate fracture repair. However, recent evidence suggests that stimulation of blood vessel formation is an oversimplified approach to support bone regeneration. This review discusses the role of vascularization during bone regeneration and delineates a phenomenon, for which we coin the term “the vascularization paradox of non-union-formation”. This view is based on the results of a variety of experimental studies that suggest that the callus tissue of non-unions is indeed densely vascularized and that pro-angiogenic mediators, such as vascular endothelial growth factor, are sufficiently expressed at the facture site. By gaining further insights into the molecular and cellular basis of non-union vascularization, it may be possible to develop more optimized treatment approaches or even prevent the non-union formation in the future.
Collapse
|
20
|
Wu F, Yang J, Ke X, Ye S, Bao Z, Yang X, Zhong C, Shen M, Xu S, Zhang L, Gou Z, Yang G. Integrating pore architectures to evaluate vascularization efficacy in silicate-based bioceramic scaffolds. Regen Biomater 2021; 9:rbab077. [PMID: 35480859 PMCID: PMC9039507 DOI: 10.1093/rb/rbab077] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 11/20/2021] [Accepted: 11/30/2021] [Indexed: 11/15/2022] Open
Abstract
Pore architecture in bioceramic scaffolds plays an important role in facilitating vascularization efficiency during bone repair or orbital reconstruction. Many investigations have explored this relationship but lack integrating pore architectural features in a scaffold, hindering optimization of architectural parameters (geometry, size and curvature) to improve vascularization and consequently clinical outcomes. To address this challenge, we have developed an integrating design strategy to fabricate different pore architectures (cube, gyroid and hexagon) with different pore dimensions (∼350, 500 and 650 μm) in the silicate-based bioceramic scaffolds via digital light processing technique. The sintered scaffolds maintained high-fidelity pore architectures similar to the printing model. The hexagon- and gyroid-pore scaffolds exhibited the highest and lowest compressive strength (from 15 to 55 MPa), respectively, but the cube-pore scaffolds showed appreciable elastic modulus. Moreover, the gyroid-pore architecture contributed on a faster ion dissolution and mass decay in vitro. It is interesting that both μCT and histological analyses indicate vascularization efficiency was challenged even in the 650-μm pore region of hexagon-pore scaffolds within 2 weeks in rabbit models, but the gyroid-pore constructs indicated appreciable blood vessel networks even in the 350-μm pore region at 2 weeks and high-density blood vessels were uniformly invaded in the 500- and 650-μm pore at 4 weeks. Angiogenesis was facilitated in the cube-pore scaffolds in comparison with the hexagon-pore ones within 4 weeks. These studies demonstrate that the continuous pore wall curvature feature in gyroid-pore architecture is an important implication for biodegradation, vascular cell migration and vessel ingrowth in porous bioceramic scaffolds.
Collapse
Affiliation(s)
- Fanghui Wu
- Department of Orthopaedics, The Third Hospital Affiliated to Wenzhou Medical University & Rui’an People’s Hospital, Rui’an 325200, China
| | - Jun Yang
- Department of Orthopaedics, The Third Hospital Affiliated to Wenzhou Medical University & Rui’an People’s Hospital, Rui’an 325200, China
| | - Xiurong Ke
- Department of Orthopaedics, The Third Hospital Affiliated to Wenzhou Medical University & Rui’an People’s Hospital, Rui’an 325200, China
| | - Shuo Ye
- Department of Orthopaedics, The Third Hospital Affiliated to Wenzhou Medical University & Rui’an People’s Hospital, Rui’an 325200, China
| | - Zhaonan Bao
- Bio-nanomaterials and Regenerative Medicine Research Division, Zhejiang-California International Nanosystem Institute, Zhejiang University, Hangzhou 310058, China
| | - Xianyan Yang
- Bio-nanomaterials and Regenerative Medicine Research Division, Zhejiang-California International Nanosystem Institute, Zhejiang University, Hangzhou 310058, China
| | - Cheng Zhong
- Department of Orthopaedics, The First Affiliated Hospital, School of Medicine of Zhejiang University, Hangzhou 310003, China
| | - Miaoda Shen
- Department of Orthopaedics, The First Affiliated Hospital, School of Medicine of Zhejiang University, Hangzhou 310003, China
| | - Sanzhong Xu
- Department of Orthopaedics, The First Affiliated Hospital, School of Medicine of Zhejiang University, Hangzhou 310003, China
| | - Lei Zhang
- Department of Orthopaedics, The Third Hospital Affiliated to Wenzhou Medical University & Rui’an People’s Hospital, Rui’an 325200, China
| | - Zhongru Gou
- Bio-nanomaterials and Regenerative Medicine Research Division, Zhejiang-California International Nanosystem Institute, Zhejiang University, Hangzhou 310058, China
| | - Guojing Yang
- Department of Orthopaedics, The Third Hospital Affiliated to Wenzhou Medical University & Rui’an People’s Hospital, Rui’an 325200, China
| |
Collapse
|
21
|
Zhang X, Wang H, Hao Z. A numerical bone regeneration model incorporating angiogenesis, considering oxygen-induced secretion of vascular endothelial growth factor and vascular remodeling. J Biomech 2021; 127:110656. [PMID: 34416529 DOI: 10.1016/j.jbiomech.2021.110656] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 05/06/2021] [Accepted: 07/25/2021] [Indexed: 02/07/2023]
Abstract
Angiogenesis is considered playing an important role in bone regeneration. Studies have shown that angiogenesis is affected by biological factors, oxygen tension, and blood flow. In this paper, we propose a bone regeneration model with angiogenesis based on the theories of mechanobiology regulation, vascular network modeling, oxygen-induced secretion of vascular endothelial growth factor (VEGF), and vascular remodeling. The results showed that this model can describe the distribution and concentration of vascular endothelial growth factor induced by oxygen tension during bone regeneration, the growth and remodeling of vascular tissue under the influence of vascular endothelial growth factor and mechanical loading, and the correspondence between vascular tissue and bone regeneration.
Collapse
Affiliation(s)
- Xuanbin Zhang
- The State Key Laboratory of Tribology, Tsinghua University, Beijing 100084, China
| | - Haosen Wang
- The State Key Laboratory of Tribology, Tsinghua University, Beijing 100084, China
| | - Zhixiu Hao
- The State Key Laboratory of Tribology, Tsinghua University, Beijing 100084, China.
| |
Collapse
|
22
|
Wang C, Ying J, Nie X, Zhou T, Xiao D, Swarnkar G, Abu-Amer Y, Guan J, Shen J. Targeting angiogenesis for fracture nonunion treatment in inflammatory disease. Bone Res 2021; 9:29. [PMID: 34099632 PMCID: PMC8184936 DOI: 10.1038/s41413-021-00150-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 01/20/2021] [Accepted: 02/01/2021] [Indexed: 02/05/2023] Open
Abstract
Atrophic fracture nonunion poses a significant clinical problem with limited therapeutic interventions. In this study, we developed a unique nonunion model with high clinical relevance using serum transfer-induced rheumatoid arthritis (RA). Arthritic mice displayed fracture nonunion with the absence of fracture callus, diminished angiogenesis and fibrotic scar tissue formation leading to the failure of biomechanical properties, representing the major manifestations of atrophic nonunion in the clinic. Mechanistically, we demonstrated that the angiogenesis defect observed in RA mice was due to the downregulation of SPP1 and CXCL12 in chondrocytes, as evidenced by the restoration of angiogenesis upon SPP1 and CXCL12 treatment in vitro. In this regard, we developed a biodegradable scaffold loaded with SPP1 and CXCL12, which displayed a beneficial effect on angiogenesis and fracture repair in mice despite the presence of inflammation. Hence, these findings strongly suggest that the sustained release of SPP1 and CXCL12 represents an effective therapeutic approach to treat impaired angiogenesis and fracture nonunion under inflammatory conditions.
Collapse
Affiliation(s)
- Cuicui Wang
- grid.4367.60000 0001 2355 7002Department of Orthopaedic Surgery, School of Medicine, Washington University, St. Louis, MO USA
| | - Jun Ying
- grid.4367.60000 0001 2355 7002Department of Orthopaedic Surgery, School of Medicine, Washington University, St. Louis, MO USA ,grid.417400.60000 0004 1799 0055Department of Orthopaedic Surgery, the First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China ,grid.417400.60000 0004 1799 0055Institute of Orthopaedics and Traumatology, the First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Xiaolei Nie
- grid.4367.60000 0001 2355 7002Department of Mechanical Engineering & Materials Science, School of Engineering, Washington University, St. Louis, MO USA
| | - Tianhong Zhou
- grid.4367.60000 0001 2355 7002Department of Mechanical Engineering & Materials Science, School of Engineering, Washington University, St. Louis, MO USA
| | - Ding Xiao
- grid.4367.60000 0001 2355 7002Department of Orthopaedic Surgery, School of Medicine, Washington University, St. Louis, MO USA
| | - Gaurav Swarnkar
- grid.4367.60000 0001 2355 7002Department of Orthopaedic Surgery, School of Medicine, Washington University, St. Louis, MO USA
| | - Yousef Abu-Amer
- grid.4367.60000 0001 2355 7002Department of Orthopaedic Surgery, School of Medicine, Washington University, St. Louis, MO USA ,grid.415840.c0000 0004 0449 6533Shriners Hospital for Children, St. Louis, MO USA
| | - Jianjun Guan
- grid.4367.60000 0001 2355 7002Department of Mechanical Engineering & Materials Science, School of Engineering, Washington University, St. Louis, MO USA
| | - Jie Shen
- grid.4367.60000 0001 2355 7002Department of Orthopaedic Surgery, School of Medicine, Washington University, St. Louis, MO USA
| |
Collapse
|
23
|
Kim J, Lee G, Chang WS, Ki SH, Park JC. Comparison and Contrast of Bone and Dentin in Genetic Disorder, Morphology and Regeneration: A Review. J Bone Metab 2021; 28:1-10. [PMID: 33730779 PMCID: PMC7973397 DOI: 10.11005/jbm.2021.28.1.1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Accepted: 12/18/2020] [Indexed: 01/08/2023] Open
Abstract
The bone and dentin have distinct healing processes. The healing process of bones is regenerative, as newly formed tissues are morphologically and functionally similar to the original bone structures. In contrast, the healing process of dentin is reparative due to its failure to replicate some of its key morphological features. In this review, we compare and contrast the healing processes of bone and dentin. We describe how distinct morphological and physiological structures of the 2 tissues translate into different signaling molecules, growth factors, and matrix protein secretion.
Collapse
Affiliation(s)
- Jaehyun Kim
- College of Dental Medicine, Columbia University, New York, USA
| | - Gayeong Lee
- College of Dental Medicine, Columbia University, New York, USA
| | - Woo Sung Chang
- College of Dental Medicine, Columbia University, New York, USA
| | - Si Hyoung Ki
- Laboratory for the Study of Regenerative Dental Medicine, Department of Oral Histology-Developmental Biology & Dental Research Institute, School of Dentistry, Seoul National University, Seoul, Korea
| | - Joo-Cheol Park
- Laboratory for the Study of Regenerative Dental Medicine, Department of Oral Histology-Developmental Biology & Dental Research Institute, School of Dentistry, Seoul National University, Seoul, Korea
| |
Collapse
|
24
|
How Surface Properties of Silica Nanoparticles Influence Structural, Microstructural and Biological Properties of Polymer Nanocomposites. MATERIALS 2021; 14:ma14040843. [PMID: 33578744 PMCID: PMC7916496 DOI: 10.3390/ma14040843] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 01/02/2021] [Accepted: 02/04/2021] [Indexed: 11/26/2022]
Abstract
The aim of this work was to study effect of the type of silica nanoparticles on the properties of nanocomposites for application in the guided bone regeneration (GBR). Two types of nanometric silica particles with different size, morphology and specific surface area (SSA) i.e., high specific surface silica (hss-SiO2) and low specific surface silica (lss-SiO2), were used as nano-fillers for a resorbable polymer matrix: poly(L-lactide-co-D,L-lactide), called PLDLA. It was shown that higher surface specific area and morphology (including pore size distribution) recorded for hss-SiO2 influences chemical activity of the nanoparticle; in addition, hydroxyl groups appeared on the surface. The nanoparticle with 10 times lower specific surface area (lss-SiO2) characterized lower chemical action. In addition, a lack of hydroxyl groups on the surface obstructed apatite nucleation (reduced zeta potential in comparison to hss-SiO2), where an apatite layer appeared already after 48 h of incubation in the simulated body fluid (SBF), and no significant changes in crystallinity of PLDLA/lss-SiO2 nanocomposite material in comparison to neat PLDLA foil were observed. The presence and type of inorganic particles in the PLDLA matrix influenced various physicochemical properties such as the wettability, and the roughness parameter note for PLDLA/lss-SiO2 increased. The results of biological investigation show that the bioactive nanocomposites with hss-SiO2 may stimulate osteoblast and fibroblast cells’proliferation and secretion of collagen type I. Additionally, both nanocomposites with the nanometric silica inducted differentiation of mesenchymal cells into osteoblasts at a proliferation stage in in vitro conditions. A higher concentration of alkaline phosphatase (ALP) was observed on the material modified with hss-SiO2 silica.
Collapse
|
25
|
Duan ZW, Lu H. Effect of Mechanical Strain on Cells Involved in Fracture Healing. Orthop Surg 2021; 13:369-375. [PMID: 33496077 PMCID: PMC7957396 DOI: 10.1111/os.12885] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 10/08/2020] [Accepted: 10/26/2020] [Indexed: 12/30/2022] Open
Abstract
Secondary fracture healing is a complex multi‐stage process in which the mechanical environment plays a key role. The use of an appropriate mechanical stimulation such as strain is conducive to tissue formation between fracture ends, thus aiding the healing process. However, if the strain is too large or too small, the biological behavior of the cells involved in bone healing will be affected, resulting in non‐union or delayed healing. In this review, we summarize the current state of knowledge regarding the effect of strain on cells that play a role in the fracture‐healing process. Overall, the related literature suggests that selection of an adequate strain promotes fracture healing through the stimulation of angiogenesis and osteogenesis, along with inhibition of osteoclast differentiation and bone resorption. However, standardized methods for the application of mechanical stimulation are lacking, and a unified consensus on the mechanism by which strain promotes cell differentiation has not yet been reached. These issues, therefore, deserve further investigation.
Collapse
Affiliation(s)
- Zheng-Wei Duan
- Department of Orthopaedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hua Lu
- Department of Orthopaedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
26
|
Partial deficiency of HIF-1α in chondrocytes effected bone repair of mandibular condylar neck. Arch Oral Biol 2020; 122:105023. [PMID: 33348208 DOI: 10.1016/j.archoralbio.2020.105023] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 11/16/2020] [Accepted: 12/02/2020] [Indexed: 11/22/2022]
Abstract
OBJECTIVES This study aimed to explore the expression of hypoxia-inducible factor 1α (HIF-1α) in chondrocytes with the healing process after unilateral mandibular condylar neck osteotomy and to verify its effect on bone repair. METHODS Models of mandibular condylar neck osteotomy were established in mice. Transgenic mice with heterozygous deficiency in HIF-1α gene in chondrocytes were used. Radiographic evaluation, quantitative reverse transcription polymerase chain reaction and histomorphometric analyses were used to compare the difference in capacities of chondrogenesis, vasifaction, osteogenesis, and bone resorption. RESULTS HIF-1α was expressed in the chondrocytes of calluses. Decreased expression of HIF-1α in chondrocytes promoted the proliferation of chondrocytes and upregulated the expression of apoptosis markers. However, the density and thickness of newly formed trabecula in transgenic mice were reduced on post-osteotomy day 28, and some expression of angiogenic, osteogenic, and osteoclastogenic markers was impaired. CONCLUSIONS These results demonstrated the importance of HIF-1α to chondrocytes and bone repair during the healing process after osteotomy of the mandibular condylar neck. Decreased HIF-1α promoted the chondrocyte proliferation, and effected endochondral ossification.
Collapse
|
27
|
Assefa F, Lim J, Kim JA, Ihn HJ, Lim S, Nam SH, Bae YC, Park EK. Secretoneurin, a Neuropeptide, Enhances Bone Regeneration in a Mouse Calvarial Bone Defect Model. Tissue Eng Regen Med 2020; 18:315-324. [PMID: 33145742 DOI: 10.1007/s13770-020-00304-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 09/07/2020] [Accepted: 09/16/2020] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND This study investigates the effects of a neuropeptide, secretoneurin (SN), on bone regeneration in an experimental mouse model. METHODS The effects of SN on cell proliferation, osteoblast marker genes expression, and mineralization were evaluated using the CCK-8 assay, quantitative reverse transcriptase polymerase chain reaction (RT-PCR), and alizarin red S staining, respectively. To examine the effects of SN on bone regeneration in vivo, bone defects were created in the calvaria of ICR mice, and 0.5 or 1 µg/ml SN was applied. New bone formation was analyzed by micro-computed tomography (micro-CT) and histology. New blood vessel formation was assessed by CD34 immunohistochemistry. RESULTS SN had no significant effect on proliferation and mineralization of MC3T3-E1 cells. However, SN partially induced the gene expression of osteoblast differentiation markers such as runt-related transcription factor 2, alkaline phosphatase, collagen type I alpha 1, and osteopontin. A significant increase of bone regeneration was observed in SN treated calvarial defects. The bone volume (BV), BV/tissue volume, trabecular thickness and trabecular number values were significantly increased in the collagen sponge plus 0.5 or 1 µg/ml SN group (p < 0.01) compared with the control group. Histologic analysis also revealed increased new bone formation in the SN-treated groups. Immunohistochemical staining of CD34 showed that the SN-treated groups contained more blood vessels compared with control in the calvarial defect area. CONCLUSION SN increases new bone and blood vessel formation in a calvarial defect site. This study suggests that SN may enhance new bone formation through its potent angiogenic activity.
Collapse
Affiliation(s)
- Freshet Assefa
- Department of Oral Pathology and Regenerative Medicine, School of Dentistry, Kyungpook National University, 2177 Dalgubeol-daero, Jung-gu, Daegu, 41940, Republic of Korea
| | - Jiwon Lim
- Department of Oral Pathology and Regenerative Medicine, School of Dentistry, Kyungpook National University, 2177 Dalgubeol-daero, Jung-gu, Daegu, 41940, Republic of Korea
| | - Ju-Ang Kim
- Department of Oral Pathology and Regenerative Medicine, School of Dentistry, Kyungpook National University, 2177 Dalgubeol-daero, Jung-gu, Daegu, 41940, Republic of Korea
| | - Hye Jung Ihn
- Cell & Matrix Research Institute, Kyungpook National University, Daegu, 41944, Republic of Korea
| | - Soomin Lim
- Department of Oral Pathology and Regenerative Medicine, School of Dentistry, Kyungpook National University, 2177 Dalgubeol-daero, Jung-gu, Daegu, 41940, Republic of Korea
| | - Sang-Hyeon Nam
- Department of Oral Pathology and Regenerative Medicine, School of Dentistry, Kyungpook National University, 2177 Dalgubeol-daero, Jung-gu, Daegu, 41940, Republic of Korea
| | - Yong Chul Bae
- Department of Oral Anatomy and Neurobiology, School of Dentistry, Kyungpook National University, Daegu, 41940, South Korea
| | - Eui Kyun Park
- Department of Oral Pathology and Regenerative Medicine, School of Dentistry, Kyungpook National University, 2177 Dalgubeol-daero, Jung-gu, Daegu, 41940, Republic of Korea.
| |
Collapse
|
28
|
Guo Q, Yang J, Chen Y, Jin X, Li Z, Wen X, Xia Q, Wang Y. Salidroside improves angiogenesis-osteogenesis coupling by regulating the HIF-1α/VEGF signalling pathway in the bone environment. Eur J Pharmacol 2020; 884:173394. [PMID: 32730833 DOI: 10.1016/j.ejphar.2020.173394] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 07/13/2020] [Accepted: 07/20/2020] [Indexed: 12/24/2022]
Abstract
Angiogenesis is essential for bone formation during skeletal development. HIF-1α and the HIF-responsive gene VEGF (vascular endothelial growth factor) are reported to be a key mechanism for coupling osteogenesis and angiogenesis. Salidroside (SAL), a major biologically active compound of Rhodiola rosea L., possesses diverse pharmacological effects. However, whether SAL can protect against bone loss via the HIF-1α/VEGF pathway, specifically by inducing angiogenesis-osteogenesis coupling in vivo, remains unknown. Therefore, in the present study, we employed primary human umbilical vein endothelial cells (HUVECs) and the permanent EA.hy926 human endothelial cell line to determine the cellular and molecular effects of SAL on vascular endothelial cells and the HIF-1α-VEGF signalling pathway in the coupling of angiogenesis-osteogenesis. The in vitro study revealed that the HUVECs and EA.hy926 cells treated with conditioned medium from osteoblast cells (MG-63 cells) treated with SAL or treated directly with SAL showed enhanced proliferation, migration and capillary structure formation. However, supplementation with an anti-VEGF antibody during the treatment of endothelial cells (ECs) significantly reversed the pro-angiogenic effect of SAL. Moreover, SAL upregulated HIF-1α expression and increased its transcriptional activity, consequently upregulating VEGF expression at the mRNA and protein levels. In addition, our in vivo analysis demonstrated that SAL can stimulate endothelial sprouting from metatarsal bones. Thus, our mechanistic study demonstrated that the pro-angiogenic effects of SAL involve HIF-1α-VEGF signalling by coordinating the coupling of angiogenesis-osteogenesis in the bone environment. Therefore, we have discovered an ideal molecule that simultaneously enhances angiogenesis and osteogenesis and thereby accelerates bone healing.
Collapse
Affiliation(s)
- Qiaoyun Guo
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; Department of Pathogenic Biology and Immunology, Logistics College of Chinese People's Armed Police Forces, Tianjin, 300309, China
| | - Jing Yang
- Department of Pathogenic Biology and Immunology, Logistics College of Chinese People's Armed Police Forces, Tianjin, 300309, China
| | - Yumeng Chen
- College of Pharmacy, Tianjin Medical University, Tianjin, 300070, China
| | - Xin Jin
- Department of Pharmacology, Logistics College of Chinese People's Armed Police Forces, Tianjin, 300309, China
| | - Zongmin Li
- Department of Pathogenic Biology and Immunology, Logistics College of Chinese People's Armed Police Forces, Tianjin, 300309, China; Department of Clinical Laboratory, Shanghai Crops Hospital of Chinese People's Armed Police Forces, Shanghai, China
| | - Xiaochang Wen
- Department of Pathogenic Biology and Immunology, Logistics College of Chinese People's Armed Police Forces, Tianjin, 300309, China
| | - Qun Xia
- Department of Orthopaedics, Characteristic Medical Center of Chinese People's Armed Police Force, Tianjin, 300162, China.
| | - Yue Wang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; Department of Pathogenic Biology and Immunology, Logistics College of Chinese People's Armed Police Forces, Tianjin, 300309, China.
| |
Collapse
|
29
|
Carlisle P, Marrs J, Gaviria L, Silliman DT, Decker JF, Brown Baer P, Guda T. Quantifying Vascular Changes Surrounding Bone Regeneration in a Porcine Mandibular Defect Using Computed Tomography. Tissue Eng Part C Methods 2020; 25:721-731. [PMID: 31850839 DOI: 10.1089/ten.tec.2019.0205] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Angiogenesis is a critical process essential for optimal bone healing. Several in vitro and in vivo systems have been previously used to elucidate some of the mechanisms involved in the process of angiogenesis, and at the same time, to test potential therapeutic agents and bioactive factors that play important roles in neovascularization. Computed tomography (CT) is a noninvasive imaging technique that has recently allowed investigators to obtain a diverse range of high-resolution, three-dimensional characterization of structures, such as bone formation within bony defects. Unfortunately, to date, angiogenesis evaluation relies primarily on histology, or ex vivo imaging and few studies have utilized CT to qualitatively and quantitatively study the vascular response during bone repair. In the current study a clinical CT-based technique was used to evaluate the effects of rhBMP-2 eluting graft treatment on soft tissue vascular architecture surrounding a large segmental bone defect model in the minipig mandible. The objective of this study was to demonstrate the efficacy of contrast-enhanced, clinical 64-slice CT technology in extracting quantitative metrics of vascular architecture over a 12-week period. The results of this study show that the presence of rhBMP-2 had a positive effect on vessel volume from 4 to 12 weeks, which was explained by a concurrent increase in vessel number, which was also significantly higher at 4 weeks for the rhBMP-2 treatment. More importantly, analysis of vessel architecture showed no changes throughout the duration of the study, indicating therapeutic safety. This study validates CT analysis as a relevant imaging method for quantitative and qualitative analysis of morphological characteristics of vascular tissue around a bone healing site. Also important, the study shows that CT technology can be used in large animal models and potentially be translated into clinical models for the development of improved methods to evaluate tissue healing and vascular adaptation processes over the course of therapy. This methodology has demonstrated sensitivity to tracking spatial and temporal changes in vascularization and has the potential to be applied to studying changes in other high-contrast tissues as well. Impact Statement Tissue engineering solutions depend on the surrounding tissue response to support regeneration. The inflammatory environment and surrounding vascular supply are critical to determining if therapies will survive, engraftment occurs, and native physiology is restored. This study for the first time evaluates the blood vessel network changes in surrounding soft tissue to a bone defect site in a large animal model, using clinically available computed tomography tools and model changes in vessel number, size, and architecture. While this study focuses on rhBMP2 delivery impacting surrounding vasculature, this validated method can be extended to studying the vascular network changes in other tissues as well.
Collapse
Affiliation(s)
- Patricia Carlisle
- Dental Trauma and Research Detachment, United States Army Institute of Surgical Research, Fort Sam Houston, San Antonio, Texas.,Prytime Medical Devices, Inc., Boerne, Texas
| | - Jeffrey Marrs
- Dental Trauma and Research Detachment, United States Army Institute of Surgical Research, Fort Sam Houston, San Antonio, Texas.,School of Dentistry, University of Texas Health Science Center at San Antonio, San Antonio, Texas
| | - Laura Gaviria
- Department of Biomedical Engineering, University of Texas at San Antonio, Texas
| | - David T Silliman
- Dental Trauma and Research Detachment, United States Army Institute of Surgical Research, Fort Sam Houston, San Antonio, Texas
| | - John F Decker
- Dental Trauma and Research Detachment, United States Army Institute of Surgical Research, Fort Sam Houston, San Antonio, Texas
| | - Pamela Brown Baer
- Dental Trauma and Research Detachment, United States Army Institute of Surgical Research, Fort Sam Houston, San Antonio, Texas.,Clinical Operations and New Product Commercialization, GenCure, San Antonio, Texas
| | - Teja Guda
- Department of Biomedical Engineering, University of Texas at San Antonio, Texas
| |
Collapse
|
30
|
Stumbras A, Kuliesius P, Darinskas A, Kubilius R, Zigmantaite V, Juodzbalys G. Bone regeneration in rabbit calvarial defects using PRGF and adipose-derived stem cells: histomorphometrical analysis. Regen Med 2020; 15:1535-1549. [PMID: 32452715 DOI: 10.2217/rme-2019-0123] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Aim: The aim of this study was to evaluate the osteogenic potential of adipose-derived stem cells (ADSCs) and to assess the influence of plasma rich in growth factors (PRGF) on bone regeneration using ADSCs. Materials & methods: Bone defects were randomly allocated to the five treatment modalities: spontaneous healing, natural bovine bone mineral (BBM), BBM loaded with PRGF, BBM loaded with ADSCs and BBM loaded with a combination of ADSCs and PRGF. Results: The PRGF significantly enhanced the biomaterial-to-bone contact. Defects treated with ADSCs and PRGF or a combination of both showed the greatest bone regeneration. Conclusion: Combining PRGF and ADSCs boosts the bone graft regenerative potential at the earliest period of healing.
Collapse
Affiliation(s)
- Arturas Stumbras
- Department of Maxillofacial Surgery, Faculty of Odontology, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Povilas Kuliesius
- Department of Maxillofacial Surgery, Faculty of Odontology, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Adas Darinskas
- Laboratory of Immunology, National Cancer Institute, Lithuania
| | - Ricardas Kubilius
- Department of Maxillofacial Surgery, Faculty of Odontology, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Vilma Zigmantaite
- Animal Research Centre, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Gintaras Juodzbalys
- Department of Maxillofacial Surgery, Faculty of Odontology, Lithuanian University of Health Sciences, Kaunas, Lithuania
| |
Collapse
|
31
|
Yang SS, Oh JM, Chun S, Kim BS, Kim CS, Lee J. Tauroursodeoxycholic acid induces angiogenic activity in endothelial cells and accelerates bone regeneration. Bone 2020; 130:115073. [PMID: 31626993 DOI: 10.1016/j.bone.2019.115073] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 07/08/2019] [Accepted: 09/17/2019] [Indexed: 12/31/2022]
Abstract
Angiogenesis is a crucial process during bone tissue regeneration. The aim of this study was to investigate the angiogenic activity and the potentiation of bone regeneration via angiogenesis using tauroursodeoxycholic acid (TUDCA) in vitro and in vivo. We investigated the effect of TUDCA on proliferation and angiogenic differentiation in human umbilical vein endothelial cells (HUVECs) and the associated signaling pathway. Proliferation was determined using crystal violet assay. Angiogenic effects were evaluated based on cell migration and tube formation. In order to explore TUDCA-signaling pathways, phosphorylation of mitogen activated protein kinase, protein kinase B (AKT), and endothelial nitric oxide synthase (eNOS) was determined using western blot. Furthermore, in vivo bone formation and angiogenesis were determined using a New Zealand outbred albino rabbit calvarial defect model, while angiogenesis and bone formation were evaluated using micro-CT and histological analysis. Our results show that TUDCA significantly increased cell proliferation. Moreover, TUDCA enhanced cell migration and tube formation in HUVECs. TUDCA increased the phosphorylation of AKT, ERK1/2, c-Jun N-terminal kinase, and eNOS. Specific inhibitors of ERK1/2 (PD98059), JNK (SP600125), and AKT (AKT1/2) inhibited the TUDCA-induced migration and tube formation, while the p38 inhibitor (SB203580) did not. The in vivo study used TUDCA to accelerate new blood vessel formation and promoted bone formation in rabbit calvarial defect model. These results indicate that TUDCA plays a critical role in enhancing the angiogenesis of endothelial cells and in vivo new bone regeneration. The use of TUDCA may contribute to the regeneration of bone tissue by improving angiogenesis.
Collapse
Affiliation(s)
- Sun-Sik Yang
- Department of Oral and Maxillofacial Surgery, College of Dentistry, Wonkwang University 77 Dunsan-ro, Seo-gu, Daejeon 302-120, Republic of Korea
| | - Jung-Mi Oh
- Department of Physiology, Chonbuk National Medical School, Jeonju-si, Jeollabuk-do 54896, Republic of Korea
| | - Sungkun Chun
- Department of Physiology, Chonbuk National Medical School, Jeonju-si, Jeollabuk-do 54896, Republic of Korea
| | - Beom-Su Kim
- Carbon Nano Convergence Technology Center for Next Generation Engineers (CNN), Chonbuk National University, 567 Baekje-daero, Deokjin-gu, Jeonju-si, Jeollabuk-do 54896, Republic of Korea.
| | - Cheol Sang Kim
- Department of Bionanosystem Engineering, Graduate School, Chonbuk National University, 567 Baekje-daero, Deokjin-gu, Jeonju-si, Jeollabuk-do 54896, Republic of Korea; Division of Mechanical Design Engineering, Chonbuk National University, Jeonju-si, Jeollabuk-do 54896, Republic of Korea.
| | - Jun Lee
- Department of Oral and Maxillofacial Surgery, College of Dentistry, Wonkwang University 77 Dunsan-ro, Seo-gu, Daejeon 302-120, Republic of Korea.
| |
Collapse
|
32
|
Implantable electrical stimulation bioreactor with liquid crystal polymer-based electrodes for enhanced bone regeneration at mandibular large defects in rabbit. Med Biol Eng Comput 2019; 58:383-399. [PMID: 31853774 DOI: 10.1007/s11517-019-02046-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2018] [Accepted: 09/05/2019] [Indexed: 02/07/2023]
Abstract
The osseous regeneration of large bone defects is still a major clinical challenge in maxillofacial and orthopedic surgery. Previous studies demonstrated that biphasic electrical stimulation (ES) stimulates bone formation; however, polyimide electrode should be removed after regeneration. This study presents an implantable electrical stimulation bioreactor with electrodes based on liquid crystal polymer (LCP), which can be permanently implanted due to excellent biocompatibility to bone tissue. The bioreactor was implanted into a critical-sized bone defect and subjected to ES for one week, where bone regeneration was evaluated four weeks after surgery using micro-CT. The effect of ES via the bioreactor was compared with a sham control group and a positive control group that received recombinant human bone morphogenetic protein (rhBMP)-2 (20 μg). New bone volume per tissue volume (BV/TV) in the ES and rhBMP-2 groups increased to 132% (p < 0.05) and 174% (p < 0.01), respectively, compared to that in the sham control group. In the histological evaluation, there was no inflammation within the bone defects and adjacent to LCP in all the groups. This study showed that the ES bioreactor with LCP electrodes could enhance bone regeneration at large bone defects, where LCP can act as a mechanically resistant outer box without inflammation. Graphical abstract To enhance bone regeneration, a bioreactor comprising collagen sponge and liquid crystal polymer-based electrode was implanted in the bone defect. Within the defect, electrical current pulses having biphasic waveform were applied from the implanted bioreactor.
Collapse
|
33
|
Streckbein P, Meier M, Kähling C, Wilbrand JF, Langguth T, Schaaf H, Howaldt HP, Streckbein R, Attia S. Donor-site Morbidity after Retromolar Bone Harvesting Using a Standardised Press Fit Cylinder Protocol. MATERIALS (BASEL, SWITZERLAND) 2019; 12:E3802. [PMID: 31752347 PMCID: PMC6887743 DOI: 10.3390/ma12223802] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/18/2019] [Revised: 09/25/2019] [Accepted: 11/15/2019] [Indexed: 11/17/2022]
Abstract
Precise fitting and immobilisation of bone transplants at the recipient site is of utmost importance for the healing process. With the help of the standardised Osseo Transfer System, the recipient site is adjusted to the graft, rather than vice versa as it is typically done. The aim of this study was to analyse donor-site morbidity after harvesting cylindrical bone grafts from the retromolar region using the Osseo Transfer System. The patient satisfaction with the surgical procedures was also evaluated. All patients treated with this standardised reconstruction method between 2006 and 2013 at the Department of Cranio-Maxillofacial Surgery, University Hospital Giessen, were included in this study. Complications were recorded and evaluated. Bone graft success and patient satisfaction were documented with a questionnaire, and then confirmed by clinical and radiological follow-up examinations. Fifty-four patients were treated and 64 harvested cylindrical autologous bone grafts were transplanted. In all cases, dental implants could be inserted after bone healing. One patient lost an implant, associated with failure of the bone graft. Six patients who were examined continued to show neurological disorders in locally limited areas. No complete or long-term damage of the inferior alveolar nerve occurred. More than 94% (n = 52) of the patients were 'very satisfied' or 'satisfied' with the results and would recommend this surgical treatment to other patients. The standardised Osseo Transfer was an effective treatment option for small and mid-sized alveolar ridge augmentations. A low donor-site morbidity rate and a high transplant success rate were verified. The Osseo Transfer System demonstrated to be a reliable surgical technique without major complications. We highly recommend this surgical augmentation procedure as a surgical treatment for local bone defects.
Collapse
Affiliation(s)
- Philipp Streckbein
- Department of Cranio-Maxillofacial Surgery, Justus-Liebig University Giessen, Klinikstr. 33, 35392 Giessen, Germany; (M.M.); (C.K.); (J.-F.W.); (T.L.); (H.S.); (H.-P.H.); (S.A.)
| | - Mathias Meier
- Department of Cranio-Maxillofacial Surgery, Justus-Liebig University Giessen, Klinikstr. 33, 35392 Giessen, Germany; (M.M.); (C.K.); (J.-F.W.); (T.L.); (H.S.); (H.-P.H.); (S.A.)
| | - Christopher Kähling
- Department of Cranio-Maxillofacial Surgery, Justus-Liebig University Giessen, Klinikstr. 33, 35392 Giessen, Germany; (M.M.); (C.K.); (J.-F.W.); (T.L.); (H.S.); (H.-P.H.); (S.A.)
| | - Jan-Falco Wilbrand
- Department of Cranio-Maxillofacial Surgery, Justus-Liebig University Giessen, Klinikstr. 33, 35392 Giessen, Germany; (M.M.); (C.K.); (J.-F.W.); (T.L.); (H.S.); (H.-P.H.); (S.A.)
| | - Tobias Langguth
- Department of Cranio-Maxillofacial Surgery, Justus-Liebig University Giessen, Klinikstr. 33, 35392 Giessen, Germany; (M.M.); (C.K.); (J.-F.W.); (T.L.); (H.S.); (H.-P.H.); (S.A.)
| | - Heidrun Schaaf
- Department of Cranio-Maxillofacial Surgery, Justus-Liebig University Giessen, Klinikstr. 33, 35392 Giessen, Germany; (M.M.); (C.K.); (J.-F.W.); (T.L.); (H.S.); (H.-P.H.); (S.A.)
| | - Hans-Peter Howaldt
- Department of Cranio-Maxillofacial Surgery, Justus-Liebig University Giessen, Klinikstr. 33, 35392 Giessen, Germany; (M.M.); (C.K.); (J.-F.W.); (T.L.); (H.S.); (H.-P.H.); (S.A.)
| | - Roland Streckbein
- IZI - Institute for Postgraduate Education in Dental Implantology, Auf dem Schafsberg, 65549 Limburg, Germany;
| | - Sameh Attia
- Department of Cranio-Maxillofacial Surgery, Justus-Liebig University Giessen, Klinikstr. 33, 35392 Giessen, Germany; (M.M.); (C.K.); (J.-F.W.); (T.L.); (H.S.); (H.-P.H.); (S.A.)
| |
Collapse
|
34
|
Zhang L, Dong Y, Xue Y, Shi J, Zhang X, Liu Y, Midgley AC, Wang S. Multifunctional Triple-Layered Composite Scaffolds Combining Platelet-Rich Fibrin Promote Bone Regeneration. ACS Biomater Sci Eng 2019; 5:6691-6702. [PMID: 33423487 DOI: 10.1021/acsbiomaterials.9b01022] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
There has been substantial progress made in the development of bone regeneration materials, driven by the deficiencies that exist in current clinical products, such as finite sources, donor site complications, and potential for disease transmission. To overcome these shortcomings, multifunctional scaffolds should be developed to integrate the relationship among osteoinduction, osteoconduction, and osseointegration. In this study, we fabricated polycaprolactone/gelatin (PG) nanofiber films by electrospinning, to act as barriers against connective tissue migration into bone defect sites; chitosan/poly (γ-glutamic acid)/hydroxyapatite (CPH) hydrogels were formed by electrostatic interaction and lyophilization, to exert osteoconduction; and platelet-rich fibrin (PRF) was extracted from rat abdominal aorta and combined with composite scaffolds, to promote bone induction through the release of growth factors. Hydrogels were immersed in simulated body fluid (SBF) for 1 month to investigate mineralization in vitro. Cytocompatibility, cell barrier effect, and osteogenic differentiation were also explored in vitro. The ability to effectively regenerate bone was analyzed by implantation of triple-layered composite scaffolds into rat calvarial defects in vivo. Size-matched hydrogel filled the defect site, and then, fresh PRF was applied to the hydrogel surface. Finally, P2G3 nanofiber films were applied and attached to the surrounding soft tissue. In short, we fabricated multifunctional triple-layered scaffolds by combining the advantages of osteoinduction, osteoconduction, and osseointegration, which could give full play to the role of PRF in bone regeneration and provide new and pragmatic concepts for bone tissue regeneration in clinical applications.
Collapse
Affiliation(s)
- Lin Zhang
- Key Laboratory of Bioactive Materials for Ministry of Education, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Yunsheng Dong
- Key Laboratory of Bioactive Materials for Ministry of Education, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Yueming Xue
- Key Laboratory of Bioactive Materials for Ministry of Education, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Jie Shi
- Key Laboratory of Bioactive Materials for Ministry of Education, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Xiangyun Zhang
- Key Laboratory of Bioactive Materials for Ministry of Education, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Yufei Liu
- Key Laboratory of Bioactive Materials for Ministry of Education, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Adam C Midgley
- Key Laboratory of Bioactive Materials for Ministry of Education, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Shufang Wang
- Key Laboratory of Bioactive Materials for Ministry of Education, College of Life Sciences, Nankai University, Tianjin 300071, China
| |
Collapse
|
35
|
Díaz E, Valle MB, Ribeiro S, Lanceros‑Mendez S, Barandiarán JM. A New Approach for the Fabrication of Cytocompatible PLLA-Magnetite Nanoparticle Composite Scaffolds. Int J Mol Sci 2019; 20:E4664. [PMID: 31547060 PMCID: PMC6801398 DOI: 10.3390/ijms20194664] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2019] [Revised: 09/10/2019] [Accepted: 09/15/2019] [Indexed: 11/24/2022] Open
Abstract
Magnetic biomimetic scaffolds of poly(L-lactide) (PLLA) and nanoparticles of magnetite (nFe3O4) are prepared in a wide ratio of compositions by lyophilization for bone regeneration. The magnetic properties, cytotoxicity, and the in vitro degradation of these porous materials are closely studied. The addition of magnetite at 50 °C was found to produce an interaction reaction between the ester groups of the PLLA and the metallic cations of the magnetite, causing the formation of complexes. This fact was confirmed by the analysis of the infrared spectroscopy and the gel permeation chromatography test results. They, respectively, showed a displacement of the absorption bands of the carbonyl group (C=O) of the PLLA and a scission of the polymer chains. The iron from the magnetite acted as a catalyser of the macromolecular scission reaction, which determines the final biomedical applications of the scaffolds-it does so because the reaction shortens the degradation process without appearing to influence its toxicity. None of the samples studied in the tests presented cytotoxicity, even at 70% magnetite concentrations.
Collapse
Affiliation(s)
- Esperanza Díaz
- Escuela de Ingeniería de Bilbao, Departamento de Ingeniería Minera, Metalúrgica y Ciencia de Materiales, Universidad del País Vasco (UPV/EHU), 48920 Portugalete, Spain
- BCMaterials, Basque Centre for Materials, Applications and Nanostructures, UPV/EHU Science Park, 48940 Leioa, Spain (J.M.B.)
| | - María Blanca Valle
- Facultad de Ciencia y Tecnología, Departamento de Electricidad y Electrónica, University of the Basque Country (UPV/EHU), Sarriena s/n, 48940 Leioa, Spain;
| | - Sylvie Ribeiro
- Centro de Física, Universidade do Minho, 4710-057 Braga, Portugal;
- Centre of Molecular and Environmental Biology (CBMA), Universidade do Minho, 4710-057 Braga, Portugal
| | - Senentxu Lanceros‑Mendez
- BCMaterials, Basque Centre for Materials, Applications and Nanostructures, UPV/EHU Science Park, 48940 Leioa, Spain (J.M.B.)
- IKERBASQUE, Basque Foundation for Science, 48013 Bilbao, Spain
| | - José Manuel Barandiarán
- BCMaterials, Basque Centre for Materials, Applications and Nanostructures, UPV/EHU Science Park, 48940 Leioa, Spain (J.M.B.)
- Facultad de Ciencia y Tecnología, Departamento de Electricidad y Electrónica, University of the Basque Country (UPV/EHU), Sarriena s/n, 48940 Leioa, Spain;
| |
Collapse
|
36
|
Korntner S, Lehner C, Gehwolf R, Wagner A, Grütz M, Kunkel N, Tempfer H, Traweger A. Limiting angiogenesis to modulate scar formation. Adv Drug Deliv Rev 2019; 146:170-189. [PMID: 29501628 DOI: 10.1016/j.addr.2018.02.010] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Revised: 01/22/2018] [Accepted: 02/26/2018] [Indexed: 02/06/2023]
Abstract
Angiogenesis, the process of new blood vessel formation from existing blood vessels, is a key aspect of virtually every repair process. During wound healing an extensive, but immature and leaky vascular plexus forms which is subsequently reduced by regression of non-functional vessels. More recent studies indicate that uncontrolled vessel growth or impaired vessel regression as a consequence of an excessive inflammatory response can impair wound healing, resulting in scarring and dysfunction. However, in order to elucidate targetable factors to promote functional tissue regeneration we need to understand the molecular and cellular underpinnings of physiological angiogenesis, ranging from induction to resolution of blood vessels. Especially for avascular tissues (e.g. cornea, tendon, ligament, cartilage, etc.), limiting rather than boosting vessel growth during wound repair potentially is beneficial to restore full tissue function and may result in favourable long-term healing outcomes.
Collapse
|
37
|
Implantable hyaluronic acid-deferoxamine conjugate prevents nonunions through stimulation of neovascularization. NPJ Regen Med 2019; 4:11. [PMID: 31123600 PMCID: PMC6529413 DOI: 10.1038/s41536-019-0072-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Accepted: 04/08/2019] [Indexed: 01/05/2023] Open
Abstract
Approximately 6.3 million fractures occur in the U.S. annually, with 5–10% resulting in debilitating nonunions. A major limitation to achieving successful bony union is impaired neovascularization. To augment fracture healing, we designed an implantable drug delivery technology containing the angiogenic stimulant, deferoxamine (DFO). DFO activates new blood vessel formation through iron chelation and upregulation of the HIF-1α pathway. However, due to its short half-life and rapid clearance, maintaining DFO at the callus site during peak fracture angiogenesis has remained challenging. To overcome these limitations, we composed an implantable formulation of DFO conjugated to hyaluronic acid (HA). This compound immobilizes DFO within the fracture callus throughout the angiogenic window, making it a high-capacity iron sponge that amplifies blood vessel formation and prevents nonunions. We investigated implanted HA-DFO’s capacity to facilitate fracture healing in the irradiated rat mandible, a model whereby nonunions routinely develop secondary to obliteration of vascularity. HA-DFO implantation significantly improved radiomorphometrics and metrics of biomechanical strength. In addition, HA-DFO treated mandibles exhibited a remarkable 91% bone union rate, representing a 3.5-fold improvement over non-treated/irradiated controls (20% bone union rate). Collectively, our work proposes a unique methodology for the targeted delivery of DFO to fracture sites in order to facilitate neovascularization. If these findings are successfully translated into clinical practice, millions of patients will benefit from the prevention of nonunions.
Collapse
|
38
|
Xu T, Luo Y, Kong F, Lv B, Zhao S, Chen J, Liu W, Cheng L, Zhou Z, Zhou Z, Huang Y, Li L, Zhao X, Qian D, Fan J, Yin G. GIT1 is critical for formation of the CD31 hiEmcn hi vessel subtype in coupling osteogenesis with angiogenesis via modulating preosteoclasts secretion of PDGF-BB. Bone 2019; 122:218-230. [PMID: 30853660 DOI: 10.1016/j.bone.2019.03.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/25/2018] [Revised: 03/06/2019] [Accepted: 03/06/2019] [Indexed: 01/08/2023]
Abstract
G protein-coupled receptor kinase 2 interacting protein-1 (GIT1) is a scaffold protein that plays a vital role in bone modeling and remodeling during osteogenesis coupled with angiogenesis. Recent studies have shown that a specialized subset of vascular endothelium strongly positive for CD31 and Endomucin (CD31hiEmcnhi) is coupled with anabolic bone formation. Based on our previous finding that GIT1 knockout (GIT1 KO) mice have impaired angiogenesis and bone formation, we hypothesized that GIT1 affects formation of the CD31hiEmcnhi vessel subtype. In the current study, GIT1 knockout (GIT1 KO) mice displayed a significant decrease in trabecular bone mass and CD31hiEmcnhi vessel number, compared to their wild-type counterparts. In the fracture healing mouse model, GIT1 KO mice contained a lower number of CD31hiEmcnhi vessels in fracture callus at days 7 and 14. However, no significant differences in the number of preosteoclasts in bone marrow, trabecular bone and callus in GIT1 KO mice were observed, compared with wild-type mice. Notably, concentrations of serum platelet-derived growth factor-BB(PDGF-BB) secreted by preosteoclasts associated with CD31hiEmcnhi vessel formation were lower in GIT1 KO mice. In addition, PDGF-BB-associated expression of phosphorylated extracellular signal-regulated kinase- 1/2 (ERK1/2) and specificity protein 1 (SP1) was significantly decreased in preosteoclasts of GIT1 KO mice. These results collectively suggest that GIT1 is a critical participant in formation of the CD31hiEmcnhi vessel subtype, highlighting a novel biologic function of this scaffold protein in preosteoclasts.
Collapse
Affiliation(s)
- Tao Xu
- Department of Orthopedic, the First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Rd., Nanjing 210029, China
| | - YongJun Luo
- Department of Orthopedic, the First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Rd., Nanjing 210029, China
| | - FanQi Kong
- Department of Orthopedic, the First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Rd., Nanjing 210029, China
| | - Bin Lv
- Department of Orthopedics, The Affiliated People's Hospital with Jiangsu University, Zhenjiang, Jiangsu Province 212000, China
| | - ShuJie Zhao
- Department of Orthopedic, the First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Rd., Nanjing 210029, China
| | - Jian Chen
- Department of Orthopedic, the First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Rd., Nanjing 210029, China
| | - Wei Liu
- Department of Orthopedic, the First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Rd., Nanjing 210029, China
| | - Lin Cheng
- Department of Orthopedic, the First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Rd., Nanjing 210029, China
| | - Zheng Zhou
- Department of Orthopedic, the First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Rd., Nanjing 210029, China
| | - ZhiMin Zhou
- Department of Orthopedic, the First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Rd., Nanjing 210029, China
| | - YiFan Huang
- Department of Orthopedic, the First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Rd., Nanjing 210029, China
| | - LinWei Li
- Department of Orthopedic, the First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Rd., Nanjing 210029, China
| | - Xuan Zhao
- Department of Orthopedic, the First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Rd., Nanjing 210029, China
| | - DingFei Qian
- Department of Orthopedic, the First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Rd., Nanjing 210029, China
| | - Jin Fan
- Department of Orthopedic, the First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Rd., Nanjing 210029, China.
| | - GuoYong Yin
- Department of Orthopedic, the First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Rd., Nanjing 210029, China.
| |
Collapse
|
39
|
Luo Y, Li D, Xie X, Kang P. Porous, lithium-doped calcium polyphosphate composite scaffolds containing vascular endothelial growth factor (VEGF)-loaded gelatin microspheres for treating glucocorticoid-induced osteonecrosis of the femoral head. Biomed Mater 2019; 14:035013. [DOI: 10.1088/1748-605x/ab0a55] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
|
40
|
Anada T, Pan CC, Stahl AM, Mori S, Fukuda J, Suzuki O, Yang Y. Vascularized Bone-Mimetic Hydrogel Constructs by 3D Bioprinting to Promote Osteogenesis and Angiogenesis. Int J Mol Sci 2019; 20:ijms20051096. [PMID: 30836606 PMCID: PMC6429349 DOI: 10.3390/ijms20051096] [Citation(s) in RCA: 96] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 02/25/2019] [Accepted: 02/27/2019] [Indexed: 12/24/2022] Open
Abstract
Bone is a highly vascularized tissue with a unique and complex structure. Long bone consists of a peripheral cortical shell containing a network of channels for vascular penetration and an inner highly vascularized bone marrow space. Bioprinting is a powerful tool to enable rapid and precise spatial patterning of cells and biomaterials. Here we developed a two-step digital light processing technique to fabricate a bone-mimetic 3D hydrogel construct based on octacalcium phosphate (OCP), spheroids of human umbilical vein endothelial cells (HUVEC), and gelatin methacrylate (GelMA) hydrogels. The bone-mimetic 3D hydrogel construct was designed to consist of a peripheral OCP-containing GelMA ring to mimic the cortical shell, and a central GelMA ring containing HUVEC spheroids to mimic the bone marrow space. We further demonstrate that OCP, which is evenly embedded in the GelMA, stimulates the osteoblastic differentiation of mesenchymal stem cells. We refined the design of a spheroid culture device to facilitate the rapid formation of a large number of HUVEC spheroids, which were embedded into different concentrations of GelMA hydrogels. It is shown that the concentration of GelMA modulates the extent of formation of the capillary-like structures originating from the HUVEC spheroids. This cell-loaded hydrogel-based bone construct with a biomimetic dual ring structure can be potentially used for bone tissue engineering.
Collapse
Affiliation(s)
- Takahisa Anada
- Soft Materials Chemistry, Department of Applied Chemistry, Institute for Materials Chemistry and Engineering, Graduate School of Engineering, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan.
- Division of Craniofacial Function Engineering, Tohoku University Graduate School of Dentistry, 4-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan.
- Department of Orthopaedic Surgery, Stanford University School of Medicine, 300 Pasteur Drive, Stanford, CA 94305, USA.
| | - Chi-Chun Pan
- Department of Orthopaedic Surgery, Stanford University School of Medicine, 300 Pasteur Drive, Stanford, CA 94305, USA.
- Department of Mechanical Engineering, Stanford University School of Engineering, Building 380, Sloan Mathematical Center, Stanford, CA 94305, USA.
| | - Alexander M Stahl
- Department of Orthopaedic Surgery, Stanford University School of Medicine, 300 Pasteur Drive, Stanford, CA 94305, USA.
- Department of Chemistry, Stanford University, Stanford, CA 94305, USA.
| | - Satomi Mori
- Graduate School of Dentistry, Tohoku University, 4-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan.
| | - Junji Fukuda
- Faculty of Engineering, Yokohama National University, 79-5 Tokiwadai, Hodogaya-ku, Yokohama 240-8501, Japan.
| | - Osamu Suzuki
- Division of Craniofacial Function Engineering, Tohoku University Graduate School of Dentistry, 4-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan.
| | - Yunzhi Yang
- Department of Orthopaedic Surgery, Stanford University School of Medicine, 300 Pasteur Drive, Stanford, CA 94305, USA.
- Department of Materials Science and Engineering, Stanford University, 300 Pasteur Drive, Stanford, CA 94305, USA.
- Department of Bioengineering, 443 Via Ortega, Stanford University School of Engineering, Stanford, CA 94305, USA.
| |
Collapse
|
41
|
Dong X, Li H, E L, Cao J, Guo B. Bioceramic akermanite enhanced vascularization and osteogenic differentiation of human induced pluripotent stem cells in 3D scaffolds in vitro and vivo. RSC Adv 2019; 9:25462-25470. [PMID: 35530104 PMCID: PMC9070079 DOI: 10.1039/c9ra02026h] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2019] [Accepted: 06/19/2019] [Indexed: 01/10/2023] Open
Abstract
A growing number of studies suggest that the modulation of cell differentiation by biomaterials is critical for tissue engineering. In previous work, we demonstrated that human induced pluripotent stem cells (iPSCs) are remarkably promising seed cells for bone tissue engineering. In addition, we found that the ionic products of akermanite (Aker) are potential inducers of osteogenic differentiation of iPSCs. Furthermore, composite scaffolds containing polymer and bioceramics have more interesting properties compared to pure bioceramic scaffolds for bone tissue engineering. The characteristic of model biomaterials in bone tissue engineering is their ability to control the osteogenic differentiation of stem cells and simultaneously induce the angiogenesis of endothelia cells. Thus, this study aimed at investigating the effects of poly(lactic-co-glycolic acid)/Aker (PLGA-Aker) composite scaffolds on angiogenic and osteogenic differentiation of human iPSCs in order to optimize the scaffold compositions. The results from Alizarin Red S staining, qRT-PCR analysis of osteogenic genes (BMP2, RUNX2, ALP, COL1 and OCN) and angiogenic genes (VEGF and CD31) demonstrated that PLGA/Aker composite scaffolds containing 10% Aker exhibited the highest stimulatory effects on the osteogenic and angiogenic differentiation of human iPSCs among all scaffolds. After the scaffolds were implanted in nu/nu mice subcutaneous pockets and calvarial defects, H&E staining, BSP immunostaining, qRT-PCR analysis and micro-CT analysis (BMD, BV/TV) indicated that PLGA + 10% Aker scaffolds enhanced the vascularization and osteogenic differentiation of human iPSCs and stimulated the repair of bone defects. Taken together, our work indicated that combining scaffolds containing silicate bioceramic Aker and human iPSCs is a promising approach for the enhancement of bone regeneration. Bioceramics akermanite enhanced vascularization and osteogenic differentiation of human iPSCs in 3D scaffolds in vitro and vivo.![]()
Collapse
Affiliation(s)
- Xixi Dong
- Stomatology Department
- General Hospital of Chinese PLA
- Beijing 100853
- China
| | - Haiyan Li
- Med-X Research Institute
- School of Biomedical Engineering
- Shanghai Jiao Tong University
- Shanghai 200030
- China
| | - Lingling E
- Stomatology Department
- General Hospital of Chinese PLA
- Beijing 100853
- China
| | - Junkai Cao
- Stomatology Department
- General Hospital of Chinese PLA
- Beijing 100853
- China
| | - Bin Guo
- Stomatology Department
- General Hospital of Chinese PLA
- Beijing 100853
- China
| |
Collapse
|
42
|
Cao C, Huang Y, Tang Q, Zhang C, Shi L, Zhao J, Hu L, Hu Z, Liu Y, Chen L. Bidirectional juxtacrine ephrinB2/Ephs signaling promotes angiogenesis of ECs and maintains self-renewal of MSCs. Biomaterials 2018; 172:1-13. [PMID: 29709731 DOI: 10.1016/j.biomaterials.2018.04.042] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Accepted: 04/21/2018] [Indexed: 12/17/2022]
Abstract
Co-transplantation of endothelial cells (ECs) and mesenchymal stem cells (MSCs) is an important strategy for repairing complex and large bone defects. However, the ways in which ECs and MSCs interact remain to be fully clarified. We found that forward ephrinB2/Ephs signaling from hBMSCs to hUVECs promoted the tube formation of hUVECs by activating the PI3K/AKT/mTOR pathway. Reverse ephrinB2/Ephs signaling from hUVECs to hBMSCs promoted the proliferation and maintenance of hBMSCs self-renewal via upregulation of OCT4, SOX2, and YAP1. Subcutaneous co-transplantation of ECs and MSCs in nude mice confirmed that forward ephrinB2/Ephs signaling could increase the cross-sectional area of blood vessels in the transplanted area, and reverse ephrinB2/Ephs signaling could maintain the self-renewal of transplanted hBMSCs in vivo. Based on these results, ephrinB2/Ephs bidirectional juxtacrine regulation between ECs and MSCs plays a pivotal role in improving the healing of bone defects by promoting angiogenesis and achieving a sufficient number of MSCs.
Collapse
Affiliation(s)
- Cen Cao
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Ying Huang
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing 100081, China
| | - Qingming Tang
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Chenguang Zhang
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Lei Shi
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing 100081, China
| | - Jiajia Zhao
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Li Hu
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Zhewen Hu
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing 100081, China
| | - Yun Liu
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing 100081, China
| | - Lili Chen
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| |
Collapse
|
43
|
Castro J, Aristizabal O, Alves E, Louzada M, Tôrres R, Viloria M, Rezende C. Biocerâmica de fosfato de cálcio nanoestruturada micro-macroporosa em grânulos de absorção rápida no preenchimento de defeito crítico em rádio de coelhos (Oryctolagus cuniculus). ARQ BRAS MED VET ZOO 2018. [DOI: 10.1590/1678-4162-9242] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
RESUMO O objetivo do presente trabalho foi avaliar, por radiografia, histologia e densitometria óssea, o efeito da HA/βTCP em grânulos de absorção rápida em defeito ósseo crítico em rádio de coelhos. Foram utilizados 35 coelhos machos, da raça Nova Zelândia, e realizou-se um defeito crítico nos rádios direito e esquerdo. Os animais foram distribuídos em GI, enxerto autólogo e GII, HA/βTCP em grânulos de absorção rápida. Avaliações radiográficas foram feitas antes da cirurgia, após, aos oito, 15, 30, 45 e 60 dias e avaliações histológicas e de densitometria. Verificou-se diferença significativa ao se comparar a densidade mineral óssea obtida ao longo do tempo de estudo. Observou-se formação de rede vascular entre os poros da biocerâmica desde o primeiro tempo de avaliação, (oito dias). Foram observados tecido ósseo primário e trabéculas em tecido ósseo preexistente a partir de 30 dias da implantação. Aos 60 dias, constatou-se presença de matriz óssea em segmentos ósseos preexistentes, caracterizando a formação óssea centrípeta. A biocerâmica HA/βTCP nanoestruturada micro-macroporosa em grânulos de absorção rápida não causa alterações microscópicas indicativas de rejeição, permite a invasão e a multiplicação celular, bem como propicia a regeneração óssea, constituindo um implante apropriado para preenchimento de falhas ósseas críticas.
Collapse
Affiliation(s)
| | | | - E.G.L. Alves
- Universidade Federal de Minas Gerais, Brazil; Universidade de Uberaba, Brazil
| | - M.J.Q. Louzada
- Universidade Estadual Paulista Júlio de Mesquita Filho, Brazil
| | | | | | | |
Collapse
|
44
|
Marrella A, Lee TY, Lee DH, Karuthedom S, Syla D, Chawla A, Khademhosseini A, Jang HL. Engineering vascularized and innervated bone biomaterials for improved skeletal tissue regeneration. MATERIALS TODAY (KIDLINGTON, ENGLAND) 2018; 21:362-376. [PMID: 30100812 PMCID: PMC6082025 DOI: 10.1016/j.mattod.2017.10.005] [Citation(s) in RCA: 163] [Impact Index Per Article: 27.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Blood vessels and nerve fibers are distributed throughout the entirety of skeletal tissue, and play important roles during bone development and fracture healing by supplying oxygen, nutrients, and cells. However, despite the successful development of bone mimetic materials that can replace damaged bone from a structural point of view, most of the available bone biomaterials often do not induce sufficient formation of blood vessels and nerves. In part, this is due to the difficulty of integrating and regulating multiple tissue types within artificial materials, which causes a gap between native skeletal tissue. Therefore, understanding the anatomy and underlying interaction mechanisms of blood vessels and nerve fibers in skeletal tissue is important to develop biomaterials that can recapitulate its complex microenvironment. In this perspective, we highlight the structure and osteogenic functions of the vascular and nervous system in bone, in a coupled manner. In addition, we discuss important design criteria for engineering vascularized, innervated, and neurovascularized bone implant materials, as well as recent advances in the development of such biomaterials. We expect that bone implant materials with neurovascularized networks can more accurately mimic native skeletal tissue and improve the regeneration of bone tissue.
Collapse
Affiliation(s)
- Alessandra Marrella
- Division of Biomedical Engineering, Department of Medicine, Biomaterials Innovation Research Center, Harvard Medical School, Brigham & Women’s Hospital, Boston, MA 02139, USA
- Division of Health Sciences & Technology, Harvard-Massachusetts Institute of Technology, Massachusetts Institute of Technology, Cambridge, MA 02139. USA
| | - Tae Yong Lee
- Division of Biomedical Engineering, Department of Medicine, Biomaterials Innovation Research Center, Harvard Medical School, Brigham & Women’s Hospital, Boston, MA 02139, USA
- Division of Health Sciences & Technology, Harvard-Massachusetts Institute of Technology, Massachusetts Institute of Technology, Cambridge, MA 02139. USA
- Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, South Korea
| | - Dong Hoon Lee
- Division of Biomedical Engineering, Department of Medicine, Biomaterials Innovation Research Center, Harvard Medical School, Brigham & Women’s Hospital, Boston, MA 02139, USA
- Division of Health Sciences & Technology, Harvard-Massachusetts Institute of Technology, Massachusetts Institute of Technology, Cambridge, MA 02139. USA
| | - Sobha Karuthedom
- Division of Biomedical Engineering, Department of Medicine, Biomaterials Innovation Research Center, Harvard Medical School, Brigham & Women’s Hospital, Boston, MA 02139, USA
- Division of Health Sciences & Technology, Harvard-Massachusetts Institute of Technology, Massachusetts Institute of Technology, Cambridge, MA 02139. USA
| | - Denata Syla
- Division of Biomedical Engineering, Department of Medicine, Biomaterials Innovation Research Center, Harvard Medical School, Brigham & Women’s Hospital, Boston, MA 02139, USA
- Division of Health Sciences & Technology, Harvard-Massachusetts Institute of Technology, Massachusetts Institute of Technology, Cambridge, MA 02139. USA
| | - Aditya Chawla
- Division of Biomedical Engineering, Department of Medicine, Biomaterials Innovation Research Center, Harvard Medical School, Brigham & Women’s Hospital, Boston, MA 02139, USA
- Division of Health Sciences & Technology, Harvard-Massachusetts Institute of Technology, Massachusetts Institute of Technology, Cambridge, MA 02139. USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Ali Khademhosseini
- Division of Biomedical Engineering, Department of Medicine, Biomaterials Innovation Research Center, Harvard Medical School, Brigham & Women’s Hospital, Boston, MA 02139, USA
- Division of Health Sciences & Technology, Harvard-Massachusetts Institute of Technology, Massachusetts Institute of Technology, Cambridge, MA 02139. USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
- Department of Bioindustrial Technologies, College of Animal Bioscience & Technology, Konkuk University, Seoul 143-701, Republic of Korea
- Department of Physics, King Abdulaziz University, Jeddah 21569, Saudi Arabia
| | - Hae Lin Jang
- Division of Biomedical Engineering, Department of Medicine, Biomaterials Innovation Research Center, Harvard Medical School, Brigham & Women’s Hospital, Boston, MA 02139, USA
- Division of Health Sciences & Technology, Harvard-Massachusetts Institute of Technology, Massachusetts Institute of Technology, Cambridge, MA 02139. USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| |
Collapse
|
45
|
Effect of Concentrated Growth Factor (CGF) on the Promotion of Osteogenesis in Bone Marrow Stromal Cells (BMSC) in vivo. Sci Rep 2018; 8:5876. [PMID: 29651154 PMCID: PMC5897572 DOI: 10.1038/s41598-018-24364-5] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Accepted: 03/27/2018] [Indexed: 12/17/2022] Open
Abstract
The therapeutic method traditionally used in bone defect reconstruction is autologous bone grafting. The most common problems affecting this type of repair approach are bone absorption and donor trauma. The approach taken in this study overcomes these problems. Bone marrow stromal cells (BMSCs) provided the crucial seed cells. Fibrin biological scaffolds were formed by combining the BMSCs with concentrated growth factor (CGF). BMSCs were isolated from Wistar rat femurs; CGF was prepared from rat heart blood. Five repair groups were created for comparative purposes: (A) CGF + BMSCs; (B) CGF; (C) collagen + BMSCs; (D) collagen; (E) blank. After three months, the rats were sacrificed, and histopathology and three-dimensional CT images produced. Bone regeneration was significantly higher in the (A) CGF + BMSC group; osteogenesis was lower in the (B) CGF and (C) collagen + BMSC groups, at very similar levels; the (D) collagen and (E) blank groups scored the lowest results. Our research suggests that combining CGF with BMSCs leads to the formation of fibrin scaffolds that have a powerful effect on osteogenesis as well as a subsidiary angiogenic effect. SEM images of the CGF scaffolds cultured with BMSCs confirmed good CGF biocompatibility. The superior osteoinductive activity of the CGF + BMSC combination makes it an excellent biomaterial for bone regeneration.
Collapse
|
46
|
Kotsougiani D, Hundepool CA, Bulstra LF, Friedrich PF, Shin AY, Bishop AT. Bone vascularized composite allotransplantation model in swine tibial defect: Evaluation of surgical angiogenesis and transplant viability. Microsurgery 2018; 39:160-166. [PMID: 29504151 DOI: 10.1002/micr.30310] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Revised: 02/02/2018] [Accepted: 02/08/2018] [Indexed: 11/10/2022]
Abstract
INTRODUCTION In prior small animal studies, we maintained vascularized bone allotransplant viability without long-term immunotherapy. Instead, an autogenous neoangiogenic circulation is created from implanted vessels, sufficient to maintain bone viability with only 2 weeks immunosupression. Blood flow is maintained despite rejection of the allogeneic vascular pedicle thereafter. We have previously described a large animal (swine) pre-clinical model, reconstructing tibial defects with vascularized tibial allotransplants. In this manuscript, autologous angiogenesis is evaluated in this model and correlated with bone viability. MATERIALS AND METHODS Allogeneic tibial segments were transplanted across a major swine leukocyte antigen mismatch. Microvascular repair of the bone VCA pedicle was combined with intraosseous implantation of an autogenous arteriovenous (AV) bundle. The bundle was ligated in group 1 (n = 4), and allowed to perfuse in group 2 (n = 4). Three-drug immunotherapy was given for 2 weeks. At 16 weeks micro-CT angiography quantified neoangiogenic vessel volume. Bone viability, rejection grade, and bone healing were analyzed. RESULTS A substantial neoangiogenic circulation developed from the implanted AV-bundle in group 2, with vessel density superior to ligated AV-bundle controls (0.11 ± 0.05 vs. 0.01 ± 0.01, P = .029). Bone allotransplant viability was also significantly enhanced by neoangiogenesis (78.7 ± 4.4% vs. 27.7 ± 5.8%, P = .028) with higher bone healing scores (21.4 ± 2.9 vs. 12.5 ± 3.7, P = .029). Ligated control tibias demonstrated disorganized bone morphology and higher local inflammation (P = .143). CONCLUSION Implantation of autogenous AV bundles into vascularized bone allotransplants resulted in the rapid formation of a neoangiogenic autogenous blood supply in a swine tibia model that maintained bone viability, improved bone healing, and minimized rejection.
Collapse
Affiliation(s)
- Dimitra Kotsougiani
- Microvascular Research Laboratory, Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota.,Department of Hand, Plastic and Reconstructive Surgery, Burn Center, BG Trauma Center Ludwigshafen, Department of Plastic Surgery, University of Heidelberg, Heidelberg, Germany
| | - Caroline A Hundepool
- Microvascular Research Laboratory, Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota
| | - Liselotte F Bulstra
- Microvascular Research Laboratory, Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota
| | - Patricia F Friedrich
- Microvascular Research Laboratory, Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota
| | - Alexander Y Shin
- Microvascular Research Laboratory, Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota
| | - Allen T Bishop
- Microvascular Research Laboratory, Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
47
|
Park J, Kim S, Kim K. Bone morphogenetic protein-2 associated multiple growth factor delivery for bone tissue regeneration. JOURNAL OF PHARMACEUTICAL INVESTIGATION 2018. [DOI: 10.1007/s40005-017-0382-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
|
48
|
Enezei HH, Ahmad A, Takeuchi K, Suzuki J, Khamis MF, Razak NHA, Rahman RA, Qabbani AA, Abdulhameed EA, Sugita Y, Maeda H, Alam MK. Osteoinductive Activity of Bone Scaffold Bioceramic Companied with Control Release of VEGF Protein Treated Dental stem cells as A New Concept for Bone Regeneration: Part II. J HARD TISSUE BIOL 2018. [DOI: 10.2485/jhtb.27.69] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Affiliation(s)
- Hamid Hammad Enezei
- Department of Oral & Maxillofacial Surgery, College of Dentistry, University of Anbar
- Department of Oral & Maxillofacial Surgery, School of Dental Sciences, Universiti Sains Malaysia Health Campus
| | - Azlina Ahmad
- Department of Biochemistry, School of Dental Sciences, Universiti Sains Malaysia Health Campus
| | - Kazuo Takeuchi
- Department of Gerodontology, School of Dentistry, Aichi Gakuin University
| | - Junji Suzuki
- Department of Oral Pathology, School of Dentistry, Aichi Gakuin University
| | - Mohd Fadhli Khamis
- Department of Oral Biology and Forensic Dentistry Unit, School of Dental Sciences, Universiti Sains Malaysia
| | - Noor Hayati Abdul Razak
- Department of Oral & Maxillofacial Surgery, School of Dental Sciences, Universiti Sains Malaysia Health Campus
| | - Roselinda Abd Rahman
- Department of Oral & Maxillofacial Surgery, School of Dental Sciences, Universiti Sains Malaysia Health Campus
| | - Ali Al Qabbani
- Department of Craniofacial Health Sciences, College of Dental Medicine, University of Sharjah
| | | | - Yoshihiko Sugita
- Department of Oral Pathology, School of Dentistry, Aichi Gakuin University
| | - Hatsuhiko Maeda
- Department of Oral Pathology, School of Dentistry, Aichi Gakuin University
| | | |
Collapse
|
49
|
Fernandez de Grado G, Keller L, Idoux-Gillet Y, Wagner Q, Musset AM, Benkirane-Jessel N, Bornert F, Offner D. Bone substitutes: a review of their characteristics, clinical use, and perspectives for large bone defects management. J Tissue Eng 2018; 9:2041731418776819. [PMID: 29899969 PMCID: PMC5990883 DOI: 10.1177/2041731418776819] [Citation(s) in RCA: 385] [Impact Index Per Article: 64.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Accepted: 04/24/2018] [Indexed: 12/13/2022] Open
Abstract
Bone replacement might have been practiced for centuries with various materials of natural origin, but had rarely met success until the late 19th century. Nowadays, many different bone substitutes can be used. They can be either derived from biological products such as demineralized bone matrix, platelet-rich plasma, hydroxyapatite, adjunction of growth factors (like bone morphogenetic protein) or synthetic such as calcium sulfate, tri-calcium phosphate ceramics, bioactive glasses, or polymer-based substitutes. All these substitutes are not suitable for every clinical use, and they have to be chosen selectively depending on their purpose. Thus, this review aims to highlight the principal characteristics of the most commonly used bone substitutes and to give some directions concerning their clinical use, as spine fusion, open-wedge tibial osteotomy, long bone fracture, oral and maxillofacial surgery, or periodontal treatments. However, the main limitations to bone substitutes use remain the management of large defects and the lack of vascularization in their central part, which is likely to appear following their utilization. In the field of bone tissue engineering, developing porous synthetic substitutes able to support a faster and a wider vascularization within their structure seems to be a promising way of research.
Collapse
Affiliation(s)
- Gabriel Fernandez de Grado
- INSERM (French National Institute of Health and Medical Research), “Regenerative Nanomedicine” laboratory, http://www.regmed.fr, UMR 1260, Faculté de Médecine, FMTS, F-67085 Strasbourg Cedex
- Université de Strasbourg, Faculté de Chirurgie Dentaire, 8 rue Ste Elisabeth, F-67000 Strasbourg
- Hôpitaux Universitaires de Strasbourg, 1 Place de l’Hôpital, F-67000 Strasbourg
| | - Laetitia Keller
- INSERM (French National Institute of Health and Medical Research), “Regenerative Nanomedicine” laboratory, http://www.regmed.fr, UMR 1260, Faculté de Médecine, FMTS, F-67085 Strasbourg Cedex
- Université de Strasbourg, Faculté de Chirurgie Dentaire, 8 rue Ste Elisabeth, F-67000 Strasbourg
| | - Ysia Idoux-Gillet
- INSERM (French National Institute of Health and Medical Research), “Regenerative Nanomedicine” laboratory, http://www.regmed.fr, UMR 1260, Faculté de Médecine, FMTS, F-67085 Strasbourg Cedex
- Université de Strasbourg, Faculté de Chirurgie Dentaire, 8 rue Ste Elisabeth, F-67000 Strasbourg
| | - Quentin Wagner
- INSERM (French National Institute of Health and Medical Research), “Regenerative Nanomedicine” laboratory, http://www.regmed.fr, UMR 1260, Faculté de Médecine, FMTS, F-67085 Strasbourg Cedex
- Université de Strasbourg, Faculté de Chirurgie Dentaire, 8 rue Ste Elisabeth, F-67000 Strasbourg
| | - Anne-Marie Musset
- INSERM (French National Institute of Health and Medical Research), “Regenerative Nanomedicine” laboratory, http://www.regmed.fr, UMR 1260, Faculté de Médecine, FMTS, F-67085 Strasbourg Cedex
- Université de Strasbourg, Faculté de Chirurgie Dentaire, 8 rue Ste Elisabeth, F-67000 Strasbourg
- Hôpitaux Universitaires de Strasbourg, 1 Place de l’Hôpital, F-67000 Strasbourg
| | - Nadia Benkirane-Jessel
- INSERM (French National Institute of Health and Medical Research), “Regenerative Nanomedicine” laboratory, http://www.regmed.fr, UMR 1260, Faculté de Médecine, FMTS, F-67085 Strasbourg Cedex
- Université de Strasbourg, Faculté de Chirurgie Dentaire, 8 rue Ste Elisabeth, F-67000 Strasbourg
| | - Fabien Bornert
- INSERM (French National Institute of Health and Medical Research), “Regenerative Nanomedicine” laboratory, http://www.regmed.fr, UMR 1260, Faculté de Médecine, FMTS, F-67085 Strasbourg Cedex
- Université de Strasbourg, Faculté de Chirurgie Dentaire, 8 rue Ste Elisabeth, F-67000 Strasbourg
- Hôpitaux Universitaires de Strasbourg, 1 Place de l’Hôpital, F-67000 Strasbourg
| | - Damien Offner
- INSERM (French National Institute of Health and Medical Research), “Regenerative Nanomedicine” laboratory, http://www.regmed.fr, UMR 1260, Faculté de Médecine, FMTS, F-67085 Strasbourg Cedex
- Université de Strasbourg, Faculté de Chirurgie Dentaire, 8 rue Ste Elisabeth, F-67000 Strasbourg
- Hôpitaux Universitaires de Strasbourg, 1 Place de l’Hôpital, F-67000 Strasbourg
| |
Collapse
|
50
|
Herath TDK, Larbi A, Teoh SH, Kirkpatrick CJ, Goh BT. Neutrophil-mediated enhancement of angiogenesis and osteogenesis in a novel triple cell co-culture model with endothelial cells and osteoblasts. J Tissue Eng Regen Med 2017; 12:e1221-e1236. [PMID: 28715156 DOI: 10.1002/term.2521] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Revised: 06/02/2017] [Accepted: 06/20/2017] [Indexed: 01/22/2023]
Abstract
Repair and regeneration of critical-sized bone defects remain a major challenge in orthopaedic and craniomaxillofacial surgery. Until now, attempts to bioengineer bone tissue have been hindered by the inability to establish proper angiogenesis and osteogenesis in the tissue construct. In the present study, we established a novel triple cell co-culture model consisting of osteoblasts, endothelial cells, and neutrophils and conducted a systematic investigation of the effects of neutrophils on angiogenesis and osteogenesis. Neutrophils significantly increased angiogenesis in the tissue construct, evidenced by the formation of microvessel-like structures with an extensive lattice-like, stable tubular network in the co-culture model. Moreover, neutrophils significantly induced the expression of pro-angiogenic markers, such as VEGF-A, CD34, EGF, and FGF-2 in a dose- and time-dependent manner. Subsequently, PCR arrays corroborated that neutrophils upregulate the important angiogenic markers and MMPs. Moreover, neutrophils also enhanced osteogenic markers, such as ALP, OCN, OPN, and COL-1 compared with the controls. As shown by the osteogenic gene arrays, neutrophils significantly regulated major osteogenic markers such as BMP2, BMP3, BMP4, BMP5, TGF-β2, RUNX2, and ECM proteins. Significantly higher mineralization was observed in triple cell co-culture compared with controls. Foregoing data indicate that the triple cell co-culture model can be used to stimulate the growth of microvasculature within a bone bioengineering construct to improve cell viability. Neutrophil-mediated enhancement of angiogenesis and osteogenesis could be a viable, clinically relevant tissue engineering strategy to obtain optimal bone growth in defect sites, in the field of oral and maxillofacial surgery.
Collapse
Affiliation(s)
| | - Anis Larbi
- Singapore Immunology Network, Agency for Science, Technology and Research, Singapore
| | - Swee Hin Teoh
- Division of Bioengineering, School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore
| | - C James Kirkpatrick
- REPAIR-Lab, Institute of Pathology, University Medical Centre, Johannes Gutenberg University, Mainz, Germany
| | | |
Collapse
|