1
|
Lu D, Zhang Y, Liang S, Li Y, Qing J, Gu L, Xu X, Wang Z, Gao X, Liu H, Zhang X, Zhou Y, Zhang P. M2 Macrophages Guide Periosteal Stromal Cell Recruitment and Initiate Bone Injury Regeneration. Biomedicines 2024; 12:1205. [PMID: 38927412 PMCID: PMC11200943 DOI: 10.3390/biomedicines12061205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 05/26/2024] [Accepted: 05/27/2024] [Indexed: 06/28/2024] Open
Abstract
The periosteum plays a critical role in bone repair and is significantly influenced by the surrounding immune microenvironment. In this study, we employed 10× single-cell RNA sequencing to create a detailed cellular atlas of the swine cranial periosteum, highlighting the cellular dynamics and interactions essential for cranial bone injury repair. We noted that such injuries lead to an increase in M2 macrophages, which are key in modulating the periosteum's immune response and driving the bone regeneration process. These macrophages actively recruit periosteal stromal cells (PSCs) by secreting Neuregulin 1 (NRG1), a crucial factor in initiating bone regeneration. This recruitment process emphasizes the critical role of PSCs in effective bone repair, positioning them as primary targets for therapeutic interventions. Our results indicate that enhancing the interaction between M2 macrophages and PSCs could significantly improve the outcomes of treatments aimed at cranial bone repair and regeneration.
Collapse
Affiliation(s)
- Dazhuang Lu
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, 22 Zhongguancun South Avenue, Haidian District, Beijing 100081, China; (D.L.); (Y.Z.); (S.L.); (Y.L.); (J.Q.); (L.G.); (X.X.); (Z.W.); (X.G.); (H.L.); (Y.Z.)
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, 22 Zhongguancun South Avenue, Haidian District, Beijing 100081, China
- Beijing Key Laboratory of Digital Stomatology, National Health Commission Key Laboratory of Digital Technology of Stomatology, 22 Zhongguancun South Avenue, Haidian District, Beijing 100081, China
- Institute of Advanced Clinical Medicine, Peking University, No.38 Xueyuan Road, Haidian District, Beijing 100191, China
| | - Yingfei Zhang
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, 22 Zhongguancun South Avenue, Haidian District, Beijing 100081, China; (D.L.); (Y.Z.); (S.L.); (Y.L.); (J.Q.); (L.G.); (X.X.); (Z.W.); (X.G.); (H.L.); (Y.Z.)
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, 22 Zhongguancun South Avenue, Haidian District, Beijing 100081, China
- Beijing Key Laboratory of Digital Stomatology, National Health Commission Key Laboratory of Digital Technology of Stomatology, 22 Zhongguancun South Avenue, Haidian District, Beijing 100081, China
- Institute of Advanced Clinical Medicine, Peking University, No.38 Xueyuan Road, Haidian District, Beijing 100191, China
| | - Shimin Liang
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, 22 Zhongguancun South Avenue, Haidian District, Beijing 100081, China; (D.L.); (Y.Z.); (S.L.); (Y.L.); (J.Q.); (L.G.); (X.X.); (Z.W.); (X.G.); (H.L.); (Y.Z.)
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, 22 Zhongguancun South Avenue, Haidian District, Beijing 100081, China
- Beijing Key Laboratory of Digital Stomatology, National Health Commission Key Laboratory of Digital Technology of Stomatology, 22 Zhongguancun South Avenue, Haidian District, Beijing 100081, China
- Institute of Advanced Clinical Medicine, Peking University, No.38 Xueyuan Road, Haidian District, Beijing 100191, China
| | - Yang Li
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, 22 Zhongguancun South Avenue, Haidian District, Beijing 100081, China; (D.L.); (Y.Z.); (S.L.); (Y.L.); (J.Q.); (L.G.); (X.X.); (Z.W.); (X.G.); (H.L.); (Y.Z.)
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, 22 Zhongguancun South Avenue, Haidian District, Beijing 100081, China
- Beijing Key Laboratory of Digital Stomatology, National Health Commission Key Laboratory of Digital Technology of Stomatology, 22 Zhongguancun South Avenue, Haidian District, Beijing 100081, China
- Institute of Advanced Clinical Medicine, Peking University, No.38 Xueyuan Road, Haidian District, Beijing 100191, China
| | - Jia Qing
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, 22 Zhongguancun South Avenue, Haidian District, Beijing 100081, China; (D.L.); (Y.Z.); (S.L.); (Y.L.); (J.Q.); (L.G.); (X.X.); (Z.W.); (X.G.); (H.L.); (Y.Z.)
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, 22 Zhongguancun South Avenue, Haidian District, Beijing 100081, China
- Beijing Key Laboratory of Digital Stomatology, National Health Commission Key Laboratory of Digital Technology of Stomatology, 22 Zhongguancun South Avenue, Haidian District, Beijing 100081, China
- Institute of Advanced Clinical Medicine, Peking University, No.38 Xueyuan Road, Haidian District, Beijing 100191, China
| | - Lanxin Gu
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, 22 Zhongguancun South Avenue, Haidian District, Beijing 100081, China; (D.L.); (Y.Z.); (S.L.); (Y.L.); (J.Q.); (L.G.); (X.X.); (Z.W.); (X.G.); (H.L.); (Y.Z.)
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, 22 Zhongguancun South Avenue, Haidian District, Beijing 100081, China
- Beijing Key Laboratory of Digital Stomatology, National Health Commission Key Laboratory of Digital Technology of Stomatology, 22 Zhongguancun South Avenue, Haidian District, Beijing 100081, China
- Institute of Advanced Clinical Medicine, Peking University, No.38 Xueyuan Road, Haidian District, Beijing 100191, China
| | - Xiuyun Xu
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, 22 Zhongguancun South Avenue, Haidian District, Beijing 100081, China; (D.L.); (Y.Z.); (S.L.); (Y.L.); (J.Q.); (L.G.); (X.X.); (Z.W.); (X.G.); (H.L.); (Y.Z.)
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, 22 Zhongguancun South Avenue, Haidian District, Beijing 100081, China
- Beijing Key Laboratory of Digital Stomatology, National Health Commission Key Laboratory of Digital Technology of Stomatology, 22 Zhongguancun South Avenue, Haidian District, Beijing 100081, China
- Institute of Advanced Clinical Medicine, Peking University, No.38 Xueyuan Road, Haidian District, Beijing 100191, China
| | - Zeying Wang
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, 22 Zhongguancun South Avenue, Haidian District, Beijing 100081, China; (D.L.); (Y.Z.); (S.L.); (Y.L.); (J.Q.); (L.G.); (X.X.); (Z.W.); (X.G.); (H.L.); (Y.Z.)
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, 22 Zhongguancun South Avenue, Haidian District, Beijing 100081, China
- Beijing Key Laboratory of Digital Stomatology, National Health Commission Key Laboratory of Digital Technology of Stomatology, 22 Zhongguancun South Avenue, Haidian District, Beijing 100081, China
- Institute of Advanced Clinical Medicine, Peking University, No.38 Xueyuan Road, Haidian District, Beijing 100191, China
| | - Xin Gao
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, 22 Zhongguancun South Avenue, Haidian District, Beijing 100081, China; (D.L.); (Y.Z.); (S.L.); (Y.L.); (J.Q.); (L.G.); (X.X.); (Z.W.); (X.G.); (H.L.); (Y.Z.)
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, 22 Zhongguancun South Avenue, Haidian District, Beijing 100081, China
- Beijing Key Laboratory of Digital Stomatology, National Health Commission Key Laboratory of Digital Technology of Stomatology, 22 Zhongguancun South Avenue, Haidian District, Beijing 100081, China
- Institute of Advanced Clinical Medicine, Peking University, No.38 Xueyuan Road, Haidian District, Beijing 100191, China
| | - Hao Liu
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, 22 Zhongguancun South Avenue, Haidian District, Beijing 100081, China; (D.L.); (Y.Z.); (S.L.); (Y.L.); (J.Q.); (L.G.); (X.X.); (Z.W.); (X.G.); (H.L.); (Y.Z.)
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, 22 Zhongguancun South Avenue, Haidian District, Beijing 100081, China
- Beijing Key Laboratory of Digital Stomatology, National Health Commission Key Laboratory of Digital Technology of Stomatology, 22 Zhongguancun South Avenue, Haidian District, Beijing 100081, China
- Institute of Advanced Clinical Medicine, Peking University, No.38 Xueyuan Road, Haidian District, Beijing 100191, China
| | - Xiao Zhang
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, 22 Zhongguancun South Avenue, Haidian District, Beijing 100081, China; (D.L.); (Y.Z.); (S.L.); (Y.L.); (J.Q.); (L.G.); (X.X.); (Z.W.); (X.G.); (H.L.); (Y.Z.)
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, 22 Zhongguancun South Avenue, Haidian District, Beijing 100081, China
- Beijing Key Laboratory of Digital Stomatology, National Health Commission Key Laboratory of Digital Technology of Stomatology, 22 Zhongguancun South Avenue, Haidian District, Beijing 100081, China
- Institute of Advanced Clinical Medicine, Peking University, No.38 Xueyuan Road, Haidian District, Beijing 100191, China
| | - Yongsheng Zhou
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, 22 Zhongguancun South Avenue, Haidian District, Beijing 100081, China; (D.L.); (Y.Z.); (S.L.); (Y.L.); (J.Q.); (L.G.); (X.X.); (Z.W.); (X.G.); (H.L.); (Y.Z.)
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, 22 Zhongguancun South Avenue, Haidian District, Beijing 100081, China
- Beijing Key Laboratory of Digital Stomatology, National Health Commission Key Laboratory of Digital Technology of Stomatology, 22 Zhongguancun South Avenue, Haidian District, Beijing 100081, China
- Institute of Advanced Clinical Medicine, Peking University, No.38 Xueyuan Road, Haidian District, Beijing 100191, China
| | - Ping Zhang
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, 22 Zhongguancun South Avenue, Haidian District, Beijing 100081, China; (D.L.); (Y.Z.); (S.L.); (Y.L.); (J.Q.); (L.G.); (X.X.); (Z.W.); (X.G.); (H.L.); (Y.Z.)
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, 22 Zhongguancun South Avenue, Haidian District, Beijing 100081, China
- Beijing Key Laboratory of Digital Stomatology, National Health Commission Key Laboratory of Digital Technology of Stomatology, 22 Zhongguancun South Avenue, Haidian District, Beijing 100081, China
- Institute of Advanced Clinical Medicine, Peking University, No.38 Xueyuan Road, Haidian District, Beijing 100191, China
| |
Collapse
|
2
|
Liu R, Jiao YR, Huang M, Zou NY, He C, Huang M, Chen KX, He WZ, Liu L, Sun YC, Xia ZY, Quarles LD, Yang HL, Wang WS, Xiao ZS, Luo XH, Li CJ. Mechanosensitive protein polycystin-1 promotes periosteal stem/progenitor cells osteochondral differentiation in fracture healing. Theranostics 2024; 14:2544-2559. [PMID: 38646641 PMCID: PMC11024844 DOI: 10.7150/thno.93269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 03/28/2024] [Indexed: 04/23/2024] Open
Abstract
Background: Mechanical forces are indispensable for bone healing, disruption of which is recognized as a contributing cause to nonunion or delayed union. However, the underlying mechanism of mechanical regulation of fracture healing is elusive. Methods: We used the lineage-tracing mouse model, conditional knockout depletion mouse model, hindlimb unloading model and single-cell RNA sequencing to analyze the crucial roles of mechanosensitive protein polycystin-1 (PC1, Pkd1) promotes periosteal stem/progenitor cells (PSPCs) osteochondral differentiation in fracture healing. Results: Our results showed that cathepsin (Ctsk)-positive PSPCs are fracture-responsive and mechanosensitive and can differentiate into osteoblasts and chondrocytes during fracture repair. We found that polycystin-1 declines markedly in PSPCs with mechanical unloading while increasing in response to mechanical stimulus. Mice with conditional depletion of Pkd1 in Ctsk+ PSPCs show impaired osteochondrogenesis, reduced cortical bone formation, delayed fracture healing, and diminished responsiveness to mechanical unloading. Mechanistically, PC1 facilitates nuclear translocation of transcriptional coactivator TAZ via PC1 C-terminal tail cleavage, enhancing osteochondral differentiation potential of PSPCs. Pharmacological intervention of the PC1-TAZ axis and promotion of TAZ nuclear translocation using Zinc01442821 enhances fracture healing and alleviates delayed union or nonunion induced by mechanical unloading. Conclusion: Our study reveals that Ctsk+ PSPCs within the callus can sense mechanical forces through the PC1-TAZ axis, targeting which represents great therapeutic potential for delayed fracture union or nonunion.
Collapse
Affiliation(s)
- Ran Liu
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan, 410008, China
| | - Yu-Rui Jiao
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan, 410008, China
| | - Mei Huang
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan, 410008, China
| | - Nan-Yu Zou
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan, 410008, China
| | - Chen He
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan, 410008, China
| | - Min Huang
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan, 410008, China
| | - Kai-Xuan Chen
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan, 410008, China
| | - Wen-Zhen He
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan, 410008, China
| | - Ling Liu
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan, 410008, China
| | - Yu-Chen Sun
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan, 410008, China
| | - Zhu-Ying Xia
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan, 410008, China
| | - L. Darryl Quarles
- Department of Medicine, University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Hai-Lin Yang
- Department of Orthopaedics, The Second Affiliated Hospital of Fuyang Normal University, Fuyang, Anhui, 236000, China
| | - Wei-Shan Wang
- Department of Orthopaedics, The First Affiliated Hospital of Shihezi University, Shihezi 832061, China
| | - Zhou-Sheng Xiao
- Department of Medicine, University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Xiang-Hang Luo
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan, 410008, China
- Key Laboratory of Aging-related Bone and Joint Diseases Prevention and Treatment, Ministry of Education, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Chang-Jun Li
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan, 410008, China
- Key Laboratory of Aging-related Bone and Joint Diseases Prevention and Treatment, Ministry of Education, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- Laboratory Animal Center, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| |
Collapse
|
3
|
Xu Y, Zhuo J, Wang Q, Xu X, He M, Zhang L, Liu Y, Wu X, Luo K, Chen Y. Site-specific periosteal cells with distinct osteogenic and angiogenic characteristics. Clin Oral Investig 2023; 27:7437-7450. [PMID: 37848582 DOI: 10.1007/s00784-023-05333-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 10/10/2023] [Indexed: 10/19/2023]
Abstract
OBJECTIVES This study aimed to investigate the site-specific characteristics of rat mandible periosteal cells (MPCs) and tibia periosteal cells (TPCs) to assess the potential application of periosteal cells (PCs) in bone tissue engineering (BTE). MATERIALS AND METHODS MPCs and TPCs were isolated and characterized. The potential of proliferation, migration, osteogenesis and adipogenesis of MPCs and TPCs were evaluated by CCK-8, scratch assay, Transwell assay, alkaline phosphatase staining and activity, Alizarin Red S staining, RT‒qPCR, and Western blot (WB) assays, respectively. Then, these cells were cocultured with human umbilical vein endothelial cells (HUVECs) to investigate their angiogenic capacity, which was assessed by scratch assay, Transwell assay, Matrigel tube formation assay, RT‒qPCR, and WB assays. RESULTS MPCs exhibited higher osteogenic potential, higher alkaline phosphatase activity, and more mineralized nodule formation, while TPCs showed a greater capability for proliferation, migration, and adipogenesis. MPCs showed higher expression of angiogenic factors, and the conditioned medium of MPCs accelerated the migration of HUVECs, while MPC- conditioned medium induced the formation of more tubular structure in HUVECs in vitro. These data suggest that compared to TPCs, MPCs exert more consequential proangiogenic effects on HUVECs. CONCLUSIONS PCs possess skeletal site-specific differences in biological characteristics. MPCs exhibit more eminent osteogenic and angiogenic potentials, which highlights the potential application of MPCs for BTE. CLINICAL RELEVANCE Autologous bone grafting as the main modality for maxillofacial bone defect repair has many limitations. Constituting an important cell type in bone repair and regeneration, MPCs show greater potential for application in BTE, which provides a promising treatment option for maxillofacial bone defect repair.
Collapse
Affiliation(s)
- Yanmei Xu
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, People's Republic of China
- Institute of Stomatology & Laboratory of Oral Tissue Engineering, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, 350002, People's Republic of China
| | - Jin Zhuo
- Xuzhou Stomatological Hospital, Affiliated Stomatological Hospital of Xuzhou Medical University, Xuzhou, 221002, People's Republic of China
| | - Qisong Wang
- Longyan First Affiliated Hospital of Fujian Medical University, Longyan, 354000, People's Republic of China
| | - Xiongcheng Xu
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, People's Republic of China
- Institute of Stomatology & Laboratory of Oral Tissue Engineering, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, 350002, People's Republic of China
| | - Mengjiao He
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, People's Republic of China
- Institute of Stomatology & Laboratory of Oral Tissue Engineering, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, 350002, People's Republic of China
| | - Lu Zhang
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, People's Republic of China
- Institute of Stomatology & Laboratory of Oral Tissue Engineering, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, 350002, People's Republic of China
| | - Yijuan Liu
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, People's Republic of China
- Institute of Stomatology & Laboratory of Oral Tissue Engineering, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, 350002, People's Republic of China
| | - Xiaohong Wu
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, People's Republic of China
- Institute of Stomatology & Laboratory of Oral Tissue Engineering, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, 350002, People's Republic of China
| | - Kai Luo
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, People's Republic of China.
- Institute of Stomatology & Laboratory of Oral Tissue Engineering, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, 350002, People's Republic of China.
| | - Yuling Chen
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, People's Republic of China.
- Institute of Stomatology & Laboratory of Oral Tissue Engineering, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, 350002, People's Republic of China.
| |
Collapse
|
4
|
Park JB, Kim I, Lee W, Kim H. Evaluation of the regenerative capacity of stem cells combined with bone graft material and collagen matrix using a rabbit calvarial defect model. J Periodontal Implant Sci 2023; 53:467-477. [PMID: 37154108 PMCID: PMC10761282 DOI: 10.5051/jpis.2204880244] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 01/26/2023] [Accepted: 02/17/2023] [Indexed: 05/10/2023] Open
Abstract
PURPOSE The purpose of this study was to evaluate the regenerative capacity of stem cells combined with bone graft material and a collagen matrix in rabbit calvarial defect models according to the type and form of the scaffolds, which included type I collagen matrix and synthetic bone. METHODS Mesenchymal stem cells (MSCs) were obtained from the periosteum of participants. Four symmetrical 6-mm-diameter circular defects were made in New Zealand white rabbits using a trephine drill. The defects were grafted with (1) group 1: synthetic bone (β-tricalcium phosphate/hydroxyapatite [β-TCP/HA]) and 1×105 MSCs; (2) group 2: collagen matrix and 1×105 MSCs; (3) group 3: β-TCP/HA, collagen matrix covering β-TCP/HA, and 1×105 MSCs; or (4) group 4: β-TCP/HA, chipped collagen matrix mixed with β-TCP/HA, and 1×105 MSCs. Cellular viability and cell migration rates were analyzed. RESULTS Uneventful healing was achieved in all areas where the defects were made at 4 weeks, and no signs of infection were identified during the healing period or at the time of retrieval. New bone formation was more evident in groups 3 and 4 than in the other groups. A densitometric analysis of the calvarium at 8 weeks post-surgery showed the highest values in group 3. CONCLUSIONS This study showed that the highest regeneration was found when the stem cells were applied to synthetic bone along with a collagen matrix.
Collapse
Affiliation(s)
- Jun-Beom Park
- Department of Periodontics, College of Medicine, The Catholic University of Korea, Seoul, Korea
- Dental Implantology, Graduate School of Clinical Dental Science, The Catholic University of Korea, Seoul, Korea
| | - InSoo Kim
- Department of Oral and Maxillofacial Surgery, College of Medicine, The Catholic University of Korea, Seoul, Korea
- Division of Oral and Maxillofacial Surgery, Department of Dentistry, Uijeongbu St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Uijeongbu, Korea.
| | - Won Lee
- Department of Oral and Maxillofacial Surgery, College of Medicine, The Catholic University of Korea, Seoul, Korea
- Division of Oral and Maxillofacial Surgery, Department of Dentistry, Uijeongbu St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Uijeongbu, Korea
| | - Heesung Kim
- Department of Oral and Maxillofacial Surgery, College of Medicine, The Catholic University of Korea, Seoul, Korea
- Division of Oral and Maxillofacial Surgery, Department of Dentistry, Uijeongbu St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Uijeongbu, Korea
- The Faculty of Liberal Arts, Eulji University, Seongnam, Korea
| |
Collapse
|
5
|
Tong YW, Chen ACY, Lei KF. Analysis of Cellular Crosstalk and Molecular Signal between Periosteum-Derived Precursor Cells and Peripheral Cells During Bone Healing Process Using a Paper-Based Osteogenesis-On-A-Chip Platform. ACS APPLIED MATERIALS & INTERFACES 2023; 15:49051-49059. [PMID: 37846857 DOI: 10.1021/acsami.3c12925] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2023]
Abstract
Periosteum-derived progenitor cells (PDPCs) are highly promising cell sources that are indispensable in the bone healing process. Adipose-derived stem cells (ADSCs) are physiologically close to periosteum tissue and release multiple growth factors to promote the bone healing process. Co-culturing PDPCs and ADSCs can construct periosteum-bone tissue microenvironments for the study of cellular crosstalk and molecular signal in the bone healing process. In the current work, a paper-based osteogenesis-on-a-chip platform was successfully developed to provide an in vitro three-dimensional coculture model. The platform was a paper substrate sandwiched between PDPC-hydrogel and ADSC-hydrogel suspensions. Cell secretion could be transferred through the paper substrate from one side to another side. Growth factors including BMP2, TGF-β, POSTN, Wnt proteins, PDGFA, and VEGFA were directly analyzed by a paper-based immunoassay. Cellular crosstalk was studied by protein expression on the paper substrate. Moreover, osteogenesis of PDPCs was investigated by examining the mRNA expressions of PDPCs after culture. Neutralizing and competitive assays were conducted to understand the correlation between growth factors secreted from ADSCs and the osteogenesis of PDPCs. In vitro periosteum-bone tissue microenvironment was established by the paper-based osteogenesis-on-a-chip platform. The proposed approach provides a promising assay of cellular crosstalk and molecular signal in 3D coculture microenvironment that may potentially lead to the development of effective bone regeneration therapy.
Collapse
Affiliation(s)
- Yun-Wen Tong
- Department of Biomedical Engineering, Chang Gung University, Taoyuan 33302, Taiwan
- Department of Orthopedic Surgery, Chang Gung Memorial Hospital, Linkou 33305, Taiwan
| | - Alvin Chao-Yu Chen
- Department of Orthopedic Surgery, Chang Gung Memorial Hospital, Linkou 33305, Taiwan
- Bone and Joint Research Center and Comprehensive Sports Medicine Center, Chang Gung Memorial Hospital, Linkou 33305, Taiwan
| | - Kin Fong Lei
- Department of Biomedical Engineering, Chang Gung University, Taoyuan 33302, Taiwan
- Department of Radiation Oncology, Chang Gung Memorial Hospital, Linkou 33305, Taiwan
- Department of Electrical & Electronic Engineering, Yonsei University, Seoul 03722, Korea
| |
Collapse
|
6
|
Xin H, Tomaskovic-Crook E, Al Maruf DSA, Cheng K, Wykes J, Manzie TGH, Wise SG, Crook JM, Clark JR. From Free Tissue Transfer to Hydrogels: A Brief Review of the Application of the Periosteum in Bone Regeneration. Gels 2023; 9:768. [PMID: 37754449 PMCID: PMC10530949 DOI: 10.3390/gels9090768] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 09/15/2023] [Accepted: 09/18/2023] [Indexed: 09/28/2023] Open
Abstract
The periosteum is a thin layer of connective tissue covering bone. It is an essential component for bone development and fracture healing. There has been considerable research exploring the application of the periosteum in bone regeneration since the 19th century. An increasing number of studies are focusing on periosteal progenitor cells found within the periosteum and the use of hydrogels as scaffold materials for periosteum engineering and guided bone development. Here, we provide an overview of the research investigating the use of the periosteum for bone repair, with consideration given to the anatomy and function of the periosteum, the importance of the cambium layer, the culture of periosteal progenitor cells, periosteum-induced ossification, periosteal perfusion, periosteum engineering, scaffold vascularization, and hydrogel-based synthetic periostea.
Collapse
Affiliation(s)
- Hai Xin
- Integrated Prosthetics and Reconstruction, Department of Head and Neck Surgery, Chris O’Brien Lifehouse, Camperdown, NSW 2050, Australia; (D.S.A.A.M.); (K.C.); (J.W.); (T.G.H.M.); (J.R.C.)
- Central Clinical School, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2050, Australia
| | - Eva Tomaskovic-Crook
- Arto Hardy Family Biomedical Innovation Hub, Chris O’Brien Lifehouse, Camperdown, NSW 2050, Australia; (E.T.-C.); (J.M.C.)
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2006, Australia;
- Intelligent Polymer Research Institute, AIIM Facility, Innovation Campus, University of Wollongong, North Wollongong, NSW 2500, Australia
| | - D S Abdullah Al Maruf
- Integrated Prosthetics and Reconstruction, Department of Head and Neck Surgery, Chris O’Brien Lifehouse, Camperdown, NSW 2050, Australia; (D.S.A.A.M.); (K.C.); (J.W.); (T.G.H.M.); (J.R.C.)
- Central Clinical School, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2050, Australia
| | - Kai Cheng
- Integrated Prosthetics and Reconstruction, Department of Head and Neck Surgery, Chris O’Brien Lifehouse, Camperdown, NSW 2050, Australia; (D.S.A.A.M.); (K.C.); (J.W.); (T.G.H.M.); (J.R.C.)
- Royal Prince Alfred Institute of Academic Surgery, Royal Prince Alfred Hospital, Sydney Local Health District, Camperdown, NSW 2050, Australia
| | - James Wykes
- Integrated Prosthetics and Reconstruction, Department of Head and Neck Surgery, Chris O’Brien Lifehouse, Camperdown, NSW 2050, Australia; (D.S.A.A.M.); (K.C.); (J.W.); (T.G.H.M.); (J.R.C.)
- Central Clinical School, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2050, Australia
| | - Timothy G. H. Manzie
- Integrated Prosthetics and Reconstruction, Department of Head and Neck Surgery, Chris O’Brien Lifehouse, Camperdown, NSW 2050, Australia; (D.S.A.A.M.); (K.C.); (J.W.); (T.G.H.M.); (J.R.C.)
| | - Steven G. Wise
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2006, Australia;
| | - Jeremy M. Crook
- Arto Hardy Family Biomedical Innovation Hub, Chris O’Brien Lifehouse, Camperdown, NSW 2050, Australia; (E.T.-C.); (J.M.C.)
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2006, Australia;
- Intelligent Polymer Research Institute, AIIM Facility, Innovation Campus, University of Wollongong, North Wollongong, NSW 2500, Australia
| | - Jonathan R. Clark
- Integrated Prosthetics and Reconstruction, Department of Head and Neck Surgery, Chris O’Brien Lifehouse, Camperdown, NSW 2050, Australia; (D.S.A.A.M.); (K.C.); (J.W.); (T.G.H.M.); (J.R.C.)
- Central Clinical School, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2050, Australia
- Royal Prince Alfred Institute of Academic Surgery, Royal Prince Alfred Hospital, Sydney Local Health District, Camperdown, NSW 2050, Australia
| |
Collapse
|
7
|
Cabaña-Muñoz ME, Pelaz Fernández MJ, Parmigiani-Cabaña JM, Parmigiani-Izquierdo JM, Merino JJ. Adult Mesenchymal Stem Cells from Oral Cavity and Surrounding Areas: Types and Biomedical Applications. Pharmaceutics 2023; 15:2109. [PMID: 37631323 PMCID: PMC10459416 DOI: 10.3390/pharmaceutics15082109] [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: 07/02/2023] [Revised: 07/28/2023] [Accepted: 08/02/2023] [Indexed: 08/27/2023] Open
Abstract
Adult mesenchymal stem cells are those obtained from the conformation of dental structures (DMSC), such as deciduous and permanent teeth and other surrounding tissues. Background: The self-renewal and differentiation capacities of these adult stem cells allow for great clinical potential. Because DMSC are cells of ectomesenchymal origin, they reveal a high capacity for complete regeneration of dental pulp, periodontal tissue, and other biomedical applications; their differentiation into other types of cells promotes repair in muscle tissue, cardiac, pancreatic, nervous, bone, cartilage, skin, and corneal tissues, among others, with a high predictability of success. Therefore, stem and progenitor cells, with their exosomes of dental origin and surrounding areas in the oral cavity due to their plasticity, are considered a fundamental pillar in medicine and regenerative dentistry. Tissue engineering (MSCs, scaffolds, and bioactive molecules) sustains and induces its multipotent and immunomodulatory effects. It is of vital importance to guarantee the safety and efficacy of the procedures designed for patients, and for this purpose, more clinical trials are needed to increase the efficacy of several pathologies. Conclusion: From a bioethical and transcendental anthropological point of view, the human person as a unique being facilitates better clinical and personalized therapy, given the higher prevalence of dental and chronic systemic diseases.
Collapse
Affiliation(s)
- María Eugenia Cabaña-Muñoz
- CIROM—Centro de Rehabilitación Oral Multidisciplinaria, 30001 Murcia, Spain; (M.E.C.-M.); (J.M.P.-C.); (J.M.P.-I.)
| | | | - José María Parmigiani-Cabaña
- CIROM—Centro de Rehabilitación Oral Multidisciplinaria, 30001 Murcia, Spain; (M.E.C.-M.); (J.M.P.-C.); (J.M.P.-I.)
| | | | - José Joaquín Merino
- Departamento de Farmacología, Farmacognosia y Botánica, Facultad de Farmacia, Universidad Complutense de Madrid (U.C.M), 28040 Madrid, Spain
| |
Collapse
|
8
|
Alarcón-Apablaza J, Prieto R, Rojas M, Fuentes R. Potential of Oral Cavity Stem Cells for Bone Regeneration: A Scoping Review. Cells 2023; 12:1392. [PMID: 37408226 PMCID: PMC10216382 DOI: 10.3390/cells12101392] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 04/30/2023] [Accepted: 05/04/2023] [Indexed: 07/07/2023] Open
Abstract
Bone loss is a common problem that ranges from small defects to large defects after trauma, surgery, or congenital malformations. The oral cavity is a rich source of mesenchymal stromal cells (MSCs). Researchers have documented their isolation and studied their osteogenic potential. Therefore, the objective of this review was to analyze and compare the potential of MSCs from the oral cavity for use in bone regeneration. METHODS A scoping review was carried out following the Preferred Reporting Items for Systematic Reviews and Meta-Analyses extension for Scoping Reviews (PRISMA-ScR) guidelines. The databases reviewed were PubMed, SCOPUS, Scientific Electronic Library Online (SciELO), and Web of Science. Studies using stem cells from the oral cavity to promote bone regeneration were included. RESULTS A total of 726 studies were found, of which 27 were selected. The MSCs used to repair bone defects were (I) dental pulp stem cells of permanent teeth, (II) stem cells derived from inflamed dental pulp, (III) stem cells from exfoliated deciduous teeth, (IV) periodontal ligament stem cells, (V) cultured autogenous periosteal cells, (VI) buccal fat pad-derived cells, and (VII) autologous bone-derived mesenchymal stem cells. Stem cells associate with scaffolds to facilitate insertion into the bone defect and to enhance bone regeneration. The biological risk and morbidity of the MSC-grafted site were minimal. Successful bone formation after MSC grafting has been shown for small defects with stem cells from the periodontal ligament and dental pulp as well as larger defects with stem cells from the periosteum, bone, and buccal fat pad. CONCLUSIONS Stem cells of maxillofacial origin are a promising alternative to treat small and large craniofacial bone defects; however, an additional scaffold complement is required for stem cell delivery.
Collapse
Affiliation(s)
- Josefa Alarcón-Apablaza
- Research Centre in Dental Sciences (CICO-UFRO), Dental School, Universidad de La Frontera, Temuco 4780000, Chile
- Doctoral Program in Morphological Sciences, Faculty of Medicine, Universidad de La Frontera, Temuco 4780000, Chile
| | - Ruth Prieto
- Department of Pediatrics and Pediatric Surgery, Faculty of Medicine, Universidad de La Frontera, Temuco 4780000, Chile
| | - Mariana Rojas
- Comparative Embryology Laboratory, Program of Anatomy and Developmental Biology, ICBM, Faculty of Medicine, Universidad de Chile, Santiago 8320000, Chile
| | - Ramón Fuentes
- Research Centre in Dental Sciences (CICO-UFRO), Dental School, Universidad de La Frontera, Temuco 4780000, Chile
- Department of Integral Adults Dentistry, Dental School, Universidad de La Frontera, Temuco 4780000, Chile
| |
Collapse
|
9
|
Adeoye AO, Hadie SNH, Munajat I, Mohd Zaharri NI, Zawawi MSF, Tuan Sharif SE, Sulaiman AR. Periosteum: Functional Anatomy and Clinical Application. MALAYSIAN JOURNAL OF MEDICINE AND HEALTH SCIENCES 2023; 19:362-374. [DOI: 10.47836/mjmhs.19.3.46] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
Periosteum is a connective tissue that envelopes the outer surface of bones and is tightly bound to the underlying bone by Sharpey’s fibers. It is composed of two layers, the outer fibrous layer and the inner cambium layer. The periosteum is densely vascularised and contains an osteoprogenitor niche that serves as a repository for bone-forming cells, which makes it an essential bone-regenerating tissue and has immensely contributed to fracture healing. Due to the high vascularity of inner cambium layer of the periosteum, periosteal transplantation has been widely used in the management of bone defects and fracture by orthopedic surgeons. Nevertheless, the use of periosteal graft in the management of bone defect is limited due to its contracted nature after being harvested. This review summarizes the current state of knowledge about the structure of periosteum, and how periosteal transplantation have been used in clinical practices, with special reference on its expansion.
Collapse
|
10
|
Leclerc K, Remark LH, Ramsukh M, Josephson AM, Palma L, Parente PEL, Sambon M, Lee S, Lopez EM, Morgani SM, Leucht P. Hox genes are crucial regulators of periosteal stem cell identity. Development 2023; 150:dev201391. [PMID: 36912250 PMCID: PMC10112919 DOI: 10.1242/dev.201391] [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: 10/19/2022] [Accepted: 02/20/2023] [Indexed: 03/14/2023]
Abstract
Periosteal stem and progenitor cells (PSPCs) are major contributors to bone maintenance and repair. Deciphering the molecular mechanisms that regulate their function is crucial for the successful generation and application of future therapeutics. Here, we pinpoint Hox transcription factors as necessary and sufficient for periosteal stem cell function. Hox genes are transcriptionally enriched in periosteal stem cells and their overexpression in more committed progenitors drives reprogramming to a naïve, self-renewing stem cell-like state. Crucially, individual Hox family members are expressed in a location-specific manner and their stem cell-promoting activity is only observed when the Hox gene is matched to the anatomical origin of the PSPC, demonstrating a role for the embryonic Hox code in adult stem cells. Finally, we demonstrate that Hoxa10 overexpression partially restores the age-related decline in fracture repair. Together, our data highlight the importance of Hox genes as key regulators of PSPC identity in skeletal homeostasis and repair.
Collapse
Affiliation(s)
- Kevin Leclerc
- Department of Orthopedic Surgery, NYU Robert I. Grossman School of Medicine, New York, NY 10016, USA
| | - Lindsey H. Remark
- Department of Orthopedic Surgery, NYU Robert I. Grossman School of Medicine, New York, NY 10016, USA
- Department of Cell Biology, NYU Robert I. Grossman School of Medicine, New York, NY 10016, USA
| | - Malissa Ramsukh
- Department of Orthopedic Surgery, NYU Robert I. Grossman School of Medicine, New York, NY 10016, USA
| | - Anne Marie Josephson
- Department of Orthopedic Surgery, NYU Robert I. Grossman School of Medicine, New York, NY 10016, USA
- Department of Cell Biology, NYU Robert I. Grossman School of Medicine, New York, NY 10016, USA
| | - Laura Palma
- Department of Orthopedic Surgery, NYU Robert I. Grossman School of Medicine, New York, NY 10016, USA
| | - Paulo E. L. Parente
- Department of Orthopedic Surgery, NYU Robert I. Grossman School of Medicine, New York, NY 10016, USA
| | - Margaux Sambon
- Department of Orthopedic Surgery, NYU Robert I. Grossman School of Medicine, New York, NY 10016, USA
| | - Sooyeon Lee
- Department of Orthopedic Surgery, NYU Robert I. Grossman School of Medicine, New York, NY 10016, USA
- Institute of Comparative Molecular Endocrinology, Ulm University, Ulm 89081, Germany
| | - Emma Muiños Lopez
- Department of Orthopedic Surgery, NYU Robert I. Grossman School of Medicine, New York, NY 10016, USA
- Cell Therapy Area, Clínica Universidad de Navarra, Pamplona 31008, Spain
| | - Sophie M. Morgani
- Department of Orthopedic Surgery, NYU Robert I. Grossman School of Medicine, New York, NY 10016, USA
| | - Philipp Leucht
- Department of Orthopedic Surgery, NYU Robert I. Grossman School of Medicine, New York, NY 10016, USA
- Department of Cell Biology, NYU Robert I. Grossman School of Medicine, New York, NY 10016, USA
| |
Collapse
|
11
|
Julien A, Perrin S, Martínez-Sarrà E, Kanagalingam A, Carvalho C, Luka M, Ménager M, Colnot C. Skeletal Stem/Progenitor Cells in Periosteum and Skeletal Muscle Share a Common Molecular Response to Bone Injury. J Bone Miner Res 2022; 37:1545-1561. [PMID: 35652423 PMCID: PMC9543664 DOI: 10.1002/jbmr.4616] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 05/19/2022] [Accepted: 05/28/2022] [Indexed: 11/07/2022]
Abstract
Bone regeneration involves skeletal stem/progenitor cells (SSPCs) recruited from bone marrow, periosteum, and adjacent skeletal muscle. To achieve bone reconstitution after injury, a coordinated cellular and molecular response is required from these cell populations. Here, we show that SSPCs from periosteum and skeletal muscle are enriched in osteochondral progenitors, and more efficiently contribute to endochondral ossification during fracture repair as compared to bone-marrow stromal cells. Single-cell RNA sequencing (RNAseq) analyses of periosteal cells reveal the cellular heterogeneity of periosteum at steady state and in response to bone fracture. Upon fracture, both periosteal and skeletal muscle SSPCs transition from a stem/progenitor to a fibrogenic state prior to chondrogenesis. This common activation pattern in periosteum and skeletal muscle SSPCs is mediated by bone morphogenetic protein (BMP) signaling. Functionally, Bmpr1a gene inactivation in platelet-derived growth factor receptor alpha (Pdgfra)-derived SSPCs impairs bone healing and decreases SSPC proliferation, migration, and osteochondral differentiation. These results uncover a coordinated molecular program driving SSPC activation in periosteum and skeletal muscle toward endochondral ossification during bone regeneration. © 2022 The Authors. Journal of Bone and Mineral Research published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Anais Julien
- Univ Paris Est Creteil, INSERM, IMRB, Creteil, France
| | - Simon Perrin
- Univ Paris Est Creteil, INSERM, IMRB, Creteil, France
| | | | | | | | - Marine Luka
- Imagine Institute, Laboratory of Inflammatory Responses and Transcriptomic Networks in Diseases, Atip-Avenir Team, INSERM UMR 1163, Université de Paris, Paris, France.,Labtech Single-Cell@Imagine, Imagine Institute, INSERM UMR 1163, Paris, France
| | - Mickaël Ménager
- Imagine Institute, Laboratory of Inflammatory Responses and Transcriptomic Networks in Diseases, Atip-Avenir Team, INSERM UMR 1163, Université de Paris, Paris, France.,Labtech Single-Cell@Imagine, Imagine Institute, INSERM UMR 1163, Paris, France
| | - Céline Colnot
- Univ Paris Est Creteil, INSERM, IMRB, Creteil, France
| |
Collapse
|
12
|
Small Intestinal Submucosa Biomimetic Periosteum Promotes Bone Regeneration. MEMBRANES 2022; 12:membranes12070719. [PMID: 35877922 PMCID: PMC9323854 DOI: 10.3390/membranes12070719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 06/10/2022] [Accepted: 07/14/2022] [Indexed: 11/30/2022]
Abstract
Background: Critical bone defects are a significant problem in clinics. The periosteum plays a vital role in bone regeneration. A tissue-engineered periosteum (TEP) has received increasing attention as a novel strategy for bone defect repairs. Methods: In this experiment, a biomimetic periosteum was fabricated by using coaxial electrospinning technology with decellularized porcine small intestinal submucosa (SIS) as the shell and polycaprolactone (PCL) as the core. In vitro, the effects of the biomimetic periosteum on Schwann cells, vascular endothelial cells, and bone marrow mesenchymal stem cells were detected by a scratch test, an EdU, a tube-forming test, and an osteogenesis test. In vivo, we used HE staining to evaluate the effect of the biomimetic periosteum on bone regeneration. Results: In vitro experiments showed that the biomimetic periosteum could significantly promote the formation of angiogenesis, osteogenesis, and repaired Schwann cells (SCs). In vivo experiments showed that the biomimetic periosteum could promote the repair of bone defects. Conclusions: The biomimetic periosteum could simulate the structural function of the periosteum and promote bone repair. This strategy may provide a promising method for the clinical treatment of skull bone defects.
Collapse
|
13
|
Histomorphometric Comparison of New Bone Formed After Maxillary Sinus Lift With Lateral and Crestal Approaches Using Periostal Mesenchymal Stem Cells and Beta-Tricalcium Phosphate: A Controlled Clinical Trial. J Craniofac Surg 2022; 33:1607-1613. [DOI: 10.1097/scs.0000000000008319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Accepted: 09/27/2021] [Indexed: 11/26/2022] Open
|
14
|
Mosaddad SA, Rasoolzade B, Namanloo RA, Azarpira N, Dortaj H. Stem cells and common biomaterials in dentistry: a review study. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2022; 33:55. [PMID: 35716227 PMCID: PMC9206624 DOI: 10.1007/s10856-022-06676-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 05/16/2022] [Indexed: 05/16/2023]
Abstract
Stem cells exist as normal cells in embryonic and adult tissues. In recent years, scientists have spared efforts to determine the role of stem cells in treating many diseases. Stem cells can self-regenerate and transform into some somatic cells. They would also have a special position in the future in various clinical fields, drug discovery, and other scientific research. Accordingly, the detection of safe and low-cost methods to obtain such cells is one of the main objectives of research. Jaw, face, and mouth tissues are the rich sources of stem cells, which more accessible than other stem cells, so stem cell and tissue engineering treatments in dentistry have received much clinical attention in recent years. This review study examines three essential elements of tissue engineering in dentistry and clinical practice, including stem cells derived from the intra- and extra-oral sources, growth factors, and scaffolds.
Collapse
Affiliation(s)
- Seyed Ali Mosaddad
- Student Research Committee, School of Dentistry, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Boshra Rasoolzade
- Student Research Committee, Department of Pediatric Dentistry, School of Dentistry, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Negar Azarpira
- Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Hengameh Dortaj
- Department of Tissue Engineering, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
15
|
Zhang N, Hu L, Cao Z, Liu X, Pan J. Periosteal Skeletal Stem Cells and Their Response to Bone Injury. Front Cell Dev Biol 2022; 10:812094. [PMID: 35399528 PMCID: PMC8987235 DOI: 10.3389/fcell.2022.812094] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 01/24/2022] [Indexed: 12/21/2022] Open
Abstract
Bone exhibits remarkable self-repair ability without fibrous scars. It is believed that the robust regenerative capacity comes from tissue-resident stem cells, such as skeletal stem cells (SSCs). Roughly, SSC has two niches: bone marrow (BM) and periosteum. BM-SSCs have been extensively studied for years. In contrast, our knowledge about periosteal SSCs (P-SSCs) is quite limited. There is abundant clinical evidence for the presence of stem cell populations within the periosteum. Researchers have even successfully cultured “stem-like” cells from the periosteum in vitro. However, due to the lack of effective markers, it is difficult to evaluate the stemness of real P-SSCs in vivo. Recently, several research teams have developed strategies for the successful identification of P-SSCs. For the first time, we can assess the stemness of P-SSCs from visual evidence. BM-SSCs and P-SSCs not only have much in common but also share distinct properties. Here, we provide an updated review of P-SSCs and their particular responses to bone injury.
Collapse
|
16
|
Rajagopal K, Ramesh S, Madhuri V. Early Addition of Parathyroid Hormone-Related Peptide Regulates the Hypertrophic Differentiation of Mesenchymal Stem Cells. Cartilage 2021; 13:143S-152S. [PMID: 31896268 PMCID: PMC8804866 DOI: 10.1177/1947603519894727] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
OBJECTIVE Chondrogenic differentiation of mesenchymal stem cells (MSCs) into hyaline cartilage is complicated by terminal hypertrophic differentiation. In growth plate, parathyroid hormone-related peptide (1-34) (PTHrP) plays a crucial role in maintaining chondrocytes in their proliferation state by counteracting the hypertrophic differentiation. This study aims to test the effect of PTHrP supplementation at different time points on chondrogenic differentiation of MSCs and assess the final quality of differentiated chondrocytes. METHODS Human periosteum and bone marrow MSCs isolated from 3 patient samples (donor unmatched) were characterized by flow cytometry and multilineage differentiation. The cells were differentiated into chondrocytes in the presence of transforming growth factor-β (TGF-β) and the PTHrP (1-34) was added from 4th or 14th day of culture. The outcome was analyzed by histology, immunohistochemistry, and gene expression. RESULTS Flow cytometry and multilineage differentiation confirmed that the cells isolated from periosteum and bone marrow exhibited the phenotype of MSCs. During chondrogenic differentiation, pellets that received PTHrP from the 4th day of culture showed a significant reduction in hypertrophic markers (COL10A1 and RUNX) than the addition of PTHrP from the 14th day and TGF-β alone treated samples. Furthermore, 4th day supplementation of PTHrP significantly improved the expression of cartilage-specific markers (COL2A1, SOX9, ACAN) in both periosteum and bone marrow-derived MSCs. Histology and immunostaining with collagen type X data corroborated the gene expression outcomes. CONCLUSION The outcome showed that supplementing PTHrP from the 4th day of chondrogenic differentiation produced better chondrocytes with less hypertrophic markers in both bone marrow and periosteal-derived MSCs.
Collapse
Affiliation(s)
- Karthikeyan Rajagopal
- Centre for Stem Cell Research, a Unit of
InStem Bengaluru, Christian Medical College, Bagayam, Vellore, Tamil Nadu,
India,Department of Paediatric Orthopaedics,
Christian Medical College, Vellore, Tamil Nadu, India
| | - Sowmya Ramesh
- Centre for Stem Cell Research, a Unit of
InStem Bengaluru, Christian Medical College, Bagayam, Vellore, Tamil Nadu,
India,Department of Paediatric Orthopaedics,
Christian Medical College, Vellore, Tamil Nadu, India
| | - Vrisha Madhuri
- Centre for Stem Cell Research, a Unit of
InStem Bengaluru, Christian Medical College, Bagayam, Vellore, Tamil Nadu,
India,Department of Paediatric Orthopaedics,
Christian Medical College, Vellore, Tamil Nadu, India,Vrisha Madhuri, Professor, Department of
Paediatric Orthopaedics, Christian Medical College, First Floor, Paul Brand
Building, Vellore 632004, Tamil Nadu, India.
| |
Collapse
|
17
|
Jeyaraman M, Muthu S, Gangadaran P, Ranjan R, Jeyaraman N, Prajwal GS, Mishra PC, Rajendran RL, Ahn BC. Osteogenic and Chondrogenic Potential of Periosteum-Derived Mesenchymal Stromal Cells: Do They Hold the Key to the Future? Pharmaceuticals (Basel) 2021; 14:ph14111133. [PMID: 34832915 PMCID: PMC8618036 DOI: 10.3390/ph14111133] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 11/02/2021] [Accepted: 11/04/2021] [Indexed: 02/05/2023] Open
Abstract
The periosteum, with its outer fibrous and inner cambium layer, lies in a dynamic environment with a niche of pluripotent stem cells for their reparative needs. The inner cambium layer is rich in mesenchymal progenitors, osteogenic progenitors, osteoblasts, and fibroblasts in a scant collagen matrix environment. Their role in union and remodeling of fracture is well known. However, the periosteum as a source of mesenchymal stem cells has not been explored in detail. Moreover, with the continuous expansion of techniques, newer insights have been acquired into the roles and regulation of these periosteal cells. From a therapeutic standpoint, the periosteum as a source of tissue engineering has gained much attraction. Apart from its role in bone repair, analysis of the bone-forming potential of periosteum-derived stem cells is lacking. Hence, this article elucidates the role of the periosteum as a potential source of mesenchymal stem cells along with their capacity for osteogenic and chondrogenic differentiation for therapeutic application in the future.
Collapse
Affiliation(s)
- Madhan Jeyaraman
- Department of Orthopaedics, School of Medical Sciences and Research, Sharda University, Greater Noida 201306, Uttar Pradesh, India; (M.J.); (R.R.)
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida 201310, Uttar Pradesh, India
- International Association of Stem Cell and Regenerative Medicine (IASRM), Greater Kailash, New Delhi 110048, Uttar Pradesh, India;
| | - Sathish Muthu
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida 201310, Uttar Pradesh, India
- International Association of Stem Cell and Regenerative Medicine (IASRM), Greater Kailash, New Delhi 110048, Uttar Pradesh, India;
- Department of Orthopaedics, Government Medical College and Hospital, Dindigul 624304, Tamil Nadu, India
- Correspondence: (S.M.); (R.L.R.); (B.-C.A.); Tel.: +82-53-420-4914 (R.L.R.); +82-53-420-5583 (B.-C.A.)
| | - Prakash Gangadaran
- BK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Department of Biomedical Sciences, School of Medicine, Kyungpook National University, Daegu 41944, Korea;
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu 41944, Korea
| | - Rajni Ranjan
- Department of Orthopaedics, School of Medical Sciences and Research, Sharda University, Greater Noida 201306, Uttar Pradesh, India; (M.J.); (R.R.)
| | - Naveen Jeyaraman
- Department of Orthopaedics, Atlas Hospitals, Tiruchirappalli 620002, Tamil Nadu, India;
| | | | - Prabhu Chandra Mishra
- International Association of Stem Cell and Regenerative Medicine (IASRM), Greater Kailash, New Delhi 110048, Uttar Pradesh, India;
| | - Ramya Lakshmi Rajendran
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu 41944, Korea
- Correspondence: (S.M.); (R.L.R.); (B.-C.A.); Tel.: +82-53-420-4914 (R.L.R.); +82-53-420-5583 (B.-C.A.)
| | - Byeong-Cheol Ahn
- BK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Department of Biomedical Sciences, School of Medicine, Kyungpook National University, Daegu 41944, Korea;
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu 41944, Korea
- Correspondence: (S.M.); (R.L.R.); (B.-C.A.); Tel.: +82-53-420-4914 (R.L.R.); +82-53-420-5583 (B.-C.A.)
| |
Collapse
|
18
|
Naung NY, Duncan WJ, De Silva RK, Coates DE. HGF/MET in osteogenic differentiation of primary human palatal periosteum-derived mesenchymal stem cells. J Oral Sci 2021; 63:341-346. [PMID: 34526445 DOI: 10.2334/josnusd.21-0164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022]
Abstract
PURPOSE This study aimed to determine expressions of hepatocyte growth factor (HGF) and MET proto-oncogene receptor tyrosine kinase (MET) in palatal periosteum (PP) and to examine the effect of HGF/MET on osteogenic differentiation of human palatal periosteum-derived mesenchymal stem cells (PD-MSCs). METHODS HGF/MET proteins in human palatal periosteum (n = 3) were localized using immunohistochemistry. PD-MSCs (n = 3) were cultured in serum-free Essential 8 (E8) medium or osteogenic medium with and without Capmatinib, a selective ATP-inhibitor of MET. HGF concentration in vitro was measured with ELISA. Relative gene expression was quantified from PD-MSCs by quantitative reverse transcription real-time polymerase chain reaction. RESULTS Immunohistochemistry detected co-localization of HGF and MET protein in PP. HGF protein levels were significantly higher (P < 0.05) in osteogenic media (day 21: 12.19 ± 8.36 ng/mL) than in E8 medium (day 21: 0.42 ± 0.72 ng/mL). MET inhibitor had a limited feedback effect on the expression profile of the osteogenic genes tested. Gene expression levels for all but three genes were comparable in serum-free and osteogenic media at all time points. CONCLUSION HGF/MET present in human PP and HGF is upregulated in vitro during osteogenesis; however the targeted pathways controlled by MET may not involve osteoblast maturation.
Collapse
Affiliation(s)
- Noel Ye Naung
- Sir John Walsh Research Institute, Faculty of Dentistry, University of Otago.,Pun Hlaing Hospitals
| | - Warwick J Duncan
- Sir John Walsh Research Institute, Faculty of Dentistry, University of Otago
| | - Rohana K De Silva
- Sir John Walsh Research Institute, Faculty of Dentistry, University of Otago
| | - Dawn E Coates
- Sir John Walsh Research Institute, Faculty of Dentistry, University of Otago
| |
Collapse
|
19
|
Kulchar RJ, Denzer BR, Chavre BM, Takegami M, Patterson J. A Review of the Use of Microparticles for Cartilage Tissue Engineering. Int J Mol Sci 2021; 22:10292. [PMID: 34638629 PMCID: PMC8508725 DOI: 10.3390/ijms221910292] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 09/17/2021] [Accepted: 09/18/2021] [Indexed: 02/06/2023] Open
Abstract
Tissue and organ failure has induced immense economic and healthcare concerns across the world. Tissue engineering is an interdisciplinary biomedical approach which aims to address the issues intrinsic to organ donation by providing an alternative strategy to tissue and organ transplantation. This review is specifically focused on cartilage tissue. Cartilage defects cannot readily regenerate, and thus research into tissue engineering approaches is relevant as a potential treatment option. Cells, scaffolds, and growth factors are three components that can be utilized to regenerate new tissue, and in particular recent advances in microparticle technology have excellent potential to revolutionize cartilage tissue regeneration. First, microspheres can be used for drug delivery by injecting them into the cartilage tissue or joint space to reduce pain and stimulate regeneration. They can also be used as controlled release systems within tissue engineering constructs. Additionally, microcarriers can act as a surface for stem cells or chondrocytes to adhere to and expand, generating large amounts of cells, which are necessary for clinically relevant cell therapies. Finally, a newer application of microparticles is to form them together into granular hydrogels to act as scaffolds for tissue engineering or to use in bioprinting. Tissue engineering has the potential to revolutionize the space of cartilage regeneration, but additional research is needed to allow for clinical translation. Microparticles are a key enabling technology in this regard.
Collapse
Affiliation(s)
- Rachel J. Kulchar
- Department of Chemistry, Princeton University, Princeton, NJ 08544, USA; (R.J.K.); (B.M.C.)
| | - Bridget R. Denzer
- Department of Chemical and Biological Engineering, Princeton University, Princeton, NJ 08544, USA;
| | - Bharvi M. Chavre
- Department of Chemistry, Princeton University, Princeton, NJ 08544, USA; (R.J.K.); (B.M.C.)
| | - Mina Takegami
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA;
| | - Jennifer Patterson
- Independent Consultant, 3000 Leuven, Belgium
- Biomaterials and Regenerative Medicine Group, IMDEA Materials Institute, 28906 Madrid, Spain
| |
Collapse
|
20
|
Influence of Human Jaw Periosteal Cells Seeded β-Tricalcium Phosphate Scaffolds on Blood Coagulation. Int J Mol Sci 2021; 22:ijms22189942. [PMID: 34576103 PMCID: PMC8467579 DOI: 10.3390/ijms22189942] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 09/08/2021] [Accepted: 09/10/2021] [Indexed: 12/13/2022] Open
Abstract
Tissue engineering offers auspicious opportunities in oral and maxillofacial surgery to heal bone defects. For this purpose, the combination of cells with stability-providing scaffolds is required. Jaw periosteal cells (JPCs) are well suited for regenerative therapies, as they are easily accessible and show strong osteogenic potential. In this study, we analyzed the influence of uncoated and polylactic-co-glycolic acid (PLGA)-coated β-tricalcium phosphate (β-TCP) scaffolds on JPC colonization and subsequent osteogenic differentiation. Furthermore, interaction with the human blood was investigated. This study demonstrated that PLGA-coated and uncoated β-TCP scaffolds can be colonized with JPCs and further differentiated into osteogenic cells. On day 15, after cell seeding, JPCs with and without osteogenic differentiation were incubated with fresh human whole blood under dynamic conditions. The activation of coagulation, complement system, inflammation, and blood cells were analyzed using ELISA and scanning electron microscopy (SEM). JPC-seeded scaffolds showed a dense cell layer and osteogenic differentiation capacity on both PLGA-coated and uncoated β-TCP scaffolds. SEM analyses showed no relevant blood cell attachment and ELISA results revealed no significant increase in most of the analyzed cell activation markers (β-thromboglobulin, Sc5B-9, polymorphonuclear (PMN)-elastase). However, a notable increase in thrombin-antithrombin III (TAT) complex levels, as well as fibrin fiber accumulation on JPC-seeded β-TCP scaffolds, was detected compared to the scaffolds without JPCs. Thus, this study demonstrated that besides the scaffold material the cells colonizing the scaffolds can also influence hemostasis, which can influence the regeneration of bone tissue.
Collapse
|
21
|
Zhu D, Zhou W, Wang Z, Wang Y, Liu M, Zhang G, Guo X, Kang X. Periostin: An Emerging Molecule With a Potential Role in Spinal Degenerative Diseases. Front Med (Lausanne) 2021; 8:694800. [PMID: 34513869 PMCID: PMC8430223 DOI: 10.3389/fmed.2021.694800] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 07/23/2021] [Indexed: 12/22/2022] Open
Abstract
Periostin, an extracellular matrix protein, is widely expressed in a variety of tissues and cells. It has many biological functions and is related to many diseases: for example, it promotes cell proliferation and differentiation in osteoblasts, which are closely related to osteoporosis, and mediates cell senescence and apoptosis in chondrocytes, which are involved in osteoarthritis. Furthermore, it also plays an important role in mediating inflammation and reconstruction during bronchial asthma, as well as in promoting bone development, reconstruction, repair, and strength. Therefore, periostin has been explored as a potential biomarker for various diseases. Recently, periostin has also been found to be expressed in intervertebral disc cells as a component of the intervertebral extracellular matrix, and to play a crucial role in the maintenance and degeneration of intervertebral discs. This article reviews the biological role of periostin in bone marrow-derived mesenchymal stem cells, osteoblasts, osteoclasts, chondrocytes, and annulus fibrosus and nucleus pulposus cells, which are closely related to spinal degenerative diseases. The study of its pathophysiological effects is of great significance for the diagnosis and treatment of spinal degeneration, although additional studies are needed.
Collapse
Affiliation(s)
- Daxue Zhu
- Lanzhou University Second Hospital, Lanzhou, China.,Orthopaedics Key Laboratory of Gansu Province, Lanzhou, China
| | - Wupin Zhou
- The 947th Army Hospital of the Chinese PLA, Kashgar, China
| | - Zhen Wang
- People's Hospital of Ningxia Hui Autonomous Region, Yinchuan, China
| | - Yidian Wang
- Lanzhou University Second Hospital, Lanzhou, China.,Orthopaedics Key Laboratory of Gansu Province, Lanzhou, China
| | - Mingqiang Liu
- Lanzhou University Second Hospital, Lanzhou, China.,Orthopaedics Key Laboratory of Gansu Province, Lanzhou, China
| | - Guangzhi Zhang
- Lanzhou University Second Hospital, Lanzhou, China.,Orthopaedics Key Laboratory of Gansu Province, Lanzhou, China
| | - Xudong Guo
- Lanzhou University Second Hospital, Lanzhou, China.,Orthopaedics Key Laboratory of Gansu Province, Lanzhou, China
| | - Xuewen Kang
- Lanzhou University Second Hospital, Lanzhou, China.,Orthopaedics Key Laboratory of Gansu Province, Lanzhou, China
| |
Collapse
|
22
|
Eichholz KF, Federici A, Riffault M, Woods I, Mahon OR, O’Driscoll L, Hoey DA. Extracellular Vesicle Functionalized Melt Electrowritten Scaffolds for Bone Tissue Engineering. ADVANCED NANOBIOMED RESEARCH 2021. [DOI: 10.1002/anbr.202100037] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Affiliation(s)
- Kian F. Eichholz
- Department of Mechanical, Aeronautical and Biomedical Engineering Materials and Surface Science Institute University of Limerick Limerick Ireland
- Trinity Centre for Biomedical Engineering Trinity Biomedical Sciences Institute Trinity College Dublin Dublin Ireland
- Department of Mechanical, Manufacturing, and Biomedical Engineering School of Engineering Trinity College Dublin Dublin Ireland
| | - Angelica Federici
- Trinity Centre for Biomedical Engineering Trinity Biomedical Sciences Institute Trinity College Dublin Dublin Ireland
- Department of Mechanical, Manufacturing, and Biomedical Engineering School of Engineering Trinity College Dublin Dublin Ireland
| | - Mathieu Riffault
- Trinity Centre for Biomedical Engineering Trinity Biomedical Sciences Institute Trinity College Dublin Dublin Ireland
- Department of Mechanical, Manufacturing, and Biomedical Engineering School of Engineering Trinity College Dublin Dublin Ireland
| | - Ian Woods
- Trinity Centre for Biomedical Engineering Trinity Biomedical Sciences Institute Trinity College Dublin Dublin Ireland
- Department of Mechanical, Manufacturing, and Biomedical Engineering School of Engineering Trinity College Dublin Dublin Ireland
| | - Olwyn R. Mahon
- Trinity Centre for Biomedical Engineering Trinity Biomedical Sciences Institute Trinity College Dublin Dublin Ireland
- Department of Mechanical, Manufacturing, and Biomedical Engineering School of Engineering Trinity College Dublin Dublin Ireland
| | - Lorraine O’Driscoll
- School of Pharmacy and Pharmaceutical Sciences Trinity College Dublin Dublin Ireland
- Trinity Biomedical Sciences Institute Trinity College Dublin Dublin Ireland
- Trinity St. James's Cancer Institute Trinity College Dublin Dublin Ireland
| | - David A. Hoey
- Department of Mechanical, Aeronautical and Biomedical Engineering Materials and Surface Science Institute University of Limerick Limerick Ireland
- Trinity Centre for Biomedical Engineering Trinity Biomedical Sciences Institute Trinity College Dublin Dublin Ireland
- Department of Mechanical, Manufacturing, and Biomedical Engineering School of Engineering Trinity College Dublin Dublin Ireland
- Advanced Materials and Bioengineering Research Centre Trinity College Dublin & RCSI Dublin Ireland
| |
Collapse
|
23
|
Gonçalves AM, Moreira A, Weber A, Williams GR, Costa PF. Osteochondral Tissue Engineering: The Potential of Electrospinning and Additive Manufacturing. Pharmaceutics 2021; 13:983. [PMID: 34209671 PMCID: PMC8309012 DOI: 10.3390/pharmaceutics13070983] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 06/22/2021] [Accepted: 06/25/2021] [Indexed: 12/14/2022] Open
Abstract
The socioeconomic impact of osteochondral (OC) damage has been increasing steadily over time in the global population, and the promise of tissue engineering in generating biomimetic tissues replicating the physiological OC environment and architecture has been falling short of its projected potential. The most recent advances in OC tissue engineering are summarised in this work, with a focus on electrospun and 3D printed biomaterials combined with stem cells and biochemical stimuli, to identify what is causing this pitfall between the bench and the patients' bedside. Even though significant progress has been achieved in electrospinning, 3D-(bio)printing, and induced pluripotent stem cell (iPSC) technologies, it is still challenging to artificially emulate the OC interface and achieve complete regeneration of bone and cartilage tissues. Their intricate architecture and the need for tight spatiotemporal control of cellular and biochemical cues hinder the attainment of long-term functional integration of tissue-engineered constructs. Moreover, this complexity and the high variability in experimental conditions used in different studies undermine the scalability and reproducibility of prospective regenerative medicine solutions. It is clear that further development of standardised, integrative, and economically viable methods regarding scaffold production, cell selection, and additional biochemical and biomechanical stimulation is likely to be the key to accelerate the clinical translation and fill the gap in OC treatment.
Collapse
Affiliation(s)
| | - Anabela Moreira
- BIOFABICS, Rua Alfredo Allen 455, 4200-135 Porto, Portugal; (A.M.G.); (A.M.)
| | - Achim Weber
- Fraunhofer Institute for Interfacial Engineering and Biotechnology IGB, Nobelstrasse 12, 70569 Stuttgart, Germany;
| | - Gareth R. Williams
- UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, UK;
| | - Pedro F. Costa
- BIOFABICS, Rua Alfredo Allen 455, 4200-135 Porto, Portugal; (A.M.G.); (A.M.)
| |
Collapse
|
24
|
Lou Y, Wang H, Ye G, Li Y, Liu C, Yu M, Ying B. Periosteal Tissue Engineering: Current Developments and Perspectives. Adv Healthc Mater 2021; 10:e2100215. [PMID: 33938636 DOI: 10.1002/adhm.202100215] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 04/18/2021] [Indexed: 12/22/2022]
Abstract
Periosteum, a highly vascularized bilayer connective tissue membrane plays an indispensable role in the repair and regeneration of bone defects. It is involved in blood supply and delivery of progenitor cells and bioactive molecules in the defect area. However, sources of natural periosteum are limited, therefore, there is a need to develop tissue-engineered periosteum (TEP) mimicking the composition, structure, and function of natural periosteum. This review explores TEP construction strategies from the following perspectives: i) different materials for constructing TEP scaffolds; ii) mechanical properties and surface topography in TEP; iii) cell-based strategies for TEP construction; and iv) TEP combined with growth factors. In addition, current challenges and future perspectives for development of TEP are discussed.
Collapse
Affiliation(s)
- Yiting Lou
- The Affiliated Hospital of Stomatology, School of Stomatology, Zhejiang University School of Medicine, Key Laboratory of Oral Biomedical Research of Zhejiang Province, 395 Yan'an road, Hangzhou, Zhejiang, 310003, China
- Department of Stomatology, The Ningbo Hospital of Zhejiang University, and Ningbo First Hospital, 59 Liuting street, Ningbo, Zhejiang, 315000, China
| | - Huiming Wang
- The Affiliated Hospital of Stomatology, School of Stomatology, Zhejiang University School of Medicine, Key Laboratory of Oral Biomedical Research of Zhejiang Province, 395 Yan'an road, Hangzhou, Zhejiang, 310003, China
| | - Guanchen Ye
- The Affiliated Hospital of Stomatology, School of Stomatology, Zhejiang University School of Medicine, Key Laboratory of Oral Biomedical Research of Zhejiang Province, 395 Yan'an road, Hangzhou, Zhejiang, 310003, China
| | - Yongzheng Li
- The Affiliated Hospital of Stomatology, School of Stomatology, Zhejiang University School of Medicine, Key Laboratory of Oral Biomedical Research of Zhejiang Province, 395 Yan'an road, Hangzhou, Zhejiang, 310003, China
| | - Chao Liu
- The Affiliated Hospital of Stomatology, School of Stomatology, Zhejiang University School of Medicine, Key Laboratory of Oral Biomedical Research of Zhejiang Province, 395 Yan'an road, Hangzhou, Zhejiang, 310003, China
| | - Mengfei Yu
- The Affiliated Hospital of Stomatology, School of Stomatology, Zhejiang University School of Medicine, Key Laboratory of Oral Biomedical Research of Zhejiang Province, 395 Yan'an road, Hangzhou, Zhejiang, 310003, China
| | - Binbin Ying
- Department of Stomatology, The Ningbo Hospital of Zhejiang University, and Ningbo First Hospital, 59 Liuting street, Ningbo, Zhejiang, 315000, China
| |
Collapse
|
25
|
Ishizuka S, Dong QN, Ngo HX, Bai Y, Sha J, Toda E, Okui T, Kanno T. Bioactive Regeneration Potential of the Newly Developed Uncalcined/Unsintered Hydroxyapatite and Poly-l-Lactide-Co-Glycolide Biomaterial in Maxillofacial Reconstructive Surgery: An In Vivo Preliminary Study. MATERIALS 2021; 14:ma14092461. [PMID: 34068558 PMCID: PMC8126161 DOI: 10.3390/ma14092461] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Revised: 05/03/2021] [Accepted: 05/07/2021] [Indexed: 12/25/2022]
Abstract
Uncalcined/unsintered hydroxyapatite (HA) and poly-l-lactide-co-glycolide (u-HA/PLLA/PGA) are novel bioresorbable bioactive materials with bone regeneration characteristics and have been used to treat mandibular defects in a rat model. However, the bone regenerative interaction with the periosteum, the inflammatory response, and the degradation of this material have not been examined. In this study, we used a rat mandible model to compare the above features in u-HA/PLLA/PGA and uncalcined/unsintered HA and poly-l-lactic acid (u-HA/PLLA). We divided 11 male Sprague–Dawley rats into 3- and 16-week groups. In each group, we assessed the characteristics of a u-HA/PLLA/PGA sheet covering the right mandibular angle and a u-HA/PLLA sheet covering the left mandibular angle in three rats each, and one rat was used as a sham control. The remaining three rats in the 16-week group were used for a degradation assessment and received both sheets of material as in the material assessment subgroup. At 3 and 16 weeks after surgery, the rats were sacrificed, and mandible specimens were subjected to micro-computed tomography, histological analysis, and immunohistochemical staining. The results indicated that the interaction between the periosteum and u-HA/PLLA/PGA material produced significantly more new bone regeneration with a lower inflammatory response and a faster resorption rate compared to u-HA/PLLA alone. These findings may indicate that this new biomaterial has ideal potential in treating maxillofacial defects of the midface and orbital regions.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Takahiro Kanno
- Correspondence: ; Tel.: +81-(0)853-20-2301; Fax: +81-(0)853-20-2299
| |
Collapse
|
26
|
Three-Dimensionally-Printed Bioactive Ceramic Scaffolds: Construct Effects on Bone Regeneration. J Craniofac Surg 2021; 32:1177-1181. [PMID: 33003153 DOI: 10.1097/scs.0000000000007146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND/PURPOSE The utilization of three-dimensionally (3D)-printed bioceramic scaffolds composed of beta-tricalcium phosphate in conjunction with dipyridamole have shown to be effective in the osteogenesis of critical bone defects in both skeletally immature and mature animals. Furthermore, previous studies have proven the dura and pericranium's osteogenic capacity in the presence of 3D-printed scaffolds; however, the effect galea aponeurotica on osteogenesis in the presence of 3D scaffolds remains unclear. METHOD/DESCRIPTION Critical-sized (11 mm) bilateral calvarial defects were created in 35-day old rabbits (n = 7). Two different 3D scaffolds were created, with one side of the calvaria being treated with a solid nonporous cap and the other with a fully porous cap. The solid cap feature was designed with the intention of preventing communication of the galea and the ossification site, while the porous cap permitted such communication. The rabbits were euthanized 8 weeks postoperatively. Calvaria were analyzed using microcomputed tomography, 3D reconstruction, and nondecalcified histologic sectioning in order assess differences in bone growth between the two types of scaffolding. RESULTS Scaffolds with the solid (nonporous) cap yielded greater percent bone volume (P = 0.012) as well as a greater percent potential bone (P = 0.001) compared with the scaffolds with a porous cap. The scaffolds with porous caps also exhibited a greater percent volume of soft tissue (P < 0.001) presence. There were no statistically significant differences detected in scaffold volume. CONCLUSION A physical barrier preventing the interaction of the galea aponeurotica with the scaffold leads to significantly increased calvarial bone regeneration in comparison with the scaffolds allowing for this interaction. The galea's interaction also leads to more soft tissue growth hindering the in growth of bone in the porous-cap scaffolds.
Collapse
|
27
|
Rajagopal K, Madhuri V. Comparing the chondrogenic potential of rabbit mesenchymal stem cells derived from the infrapatellar fat pad, periosteum & bone marrow. Indian J Med Res 2021; 154:732-742. [PMID: 35532591 PMCID: PMC9210523 DOI: 10.4103/ijmr.ijmr_93_19] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Background & objectives: Rabbit model is commonly used to demonstrate the proof of concept in cartilage tissue engineering. However, limited studies have attempted to find an ideal source of rabbit mesenchymal stem cells (MSCs) for cartilage repair. This study aimed to compare the in vitro chondrogenic potential of rabbit MSCs isolated from three sources namely infrapatellar fat pad (IFP), periosteum (P) and bone marrow (BM). Methods: Rabbit MSCs from three sources were isolated and characterized using flow cytometry and multi-lineage differentiation assay. Cell proliferation was assessed using trypan blue dye exclusion test; in vitro chondrogenic potential was evaluated by histology and gene expression and the outcomes were compared amongst the three MSC sources. Results: MSCs from three sources shared similar morphology and expressed >99 per cent positive for CD44 and CD81 and <3 per cent positive for negative markers CD34, CD90 and human leukocyte antigen – DR isotype (HLA-DR). The BM-MSCs and IFP-MSCs showed significantly higher cell proliferation (P<0.001) than the P-MSCs from passage 4. Histologically, BM-MSCs formed a thicker cartilage pellet (P<0.01) with abundant matrix deposition than IFP and P-MSCs during chondrogenic differentiation. The collagen type 2 staining was significantly (P<0.05) higher in BM-MSCs than the other two sources. These outcomes were further confirmed by gene expression, where the BM-MSCs demonstrated significantly higher expression (P<0.01) of cartilage-specific markers (COL2A1, SOX9 and ACAN) with less hypertrophy. Interpretation & conclusions: This study demonstrated that BM-MSCs had superior chondrogenic potential and generated better cartilage than IFP and P-MSCs in rabbits. Thus, BM-MSCs remain a promising candidate for rabbit articular cartilage regeneration.
Collapse
Affiliation(s)
- Karthikeyan Rajagopal
- Department of Paediatric Orthopaedic; Centre for Stem Cell Research, Christian Medical College, Vellore, Tamil Nadu, India
| | - Vrisha Madhuri
- Department of Paediatric Orthopaedic; Centre for Stem Cell Research, Christian Medical College, Vellore, Tamil Nadu, India
| |
Collapse
|
28
|
The Novel Membrane-Type Micro-system to Assess the Bonus Effect of Physiological and Physical Stimuli on Bone Regeneration. BIOCHIP JOURNAL 2021. [DOI: 10.1007/s13206-021-00023-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
29
|
Abstract
This chapter describes the methods of isolation of mouse periosteal progenitor cells. There are three basic methods utilized. The bone grafting method was developed utilizing the fracture healing process to expand the progenitor populations. Bone capping methods requires enzymatic digestion and purification of cells from the native periosteum, while the Egression/Explant method requires the least manipulation with placement of cortical bone fragments with attached periosteum in a culture dish. Various cell surface antibodies have been employed over the years to characterize periosteum derived progenitor cells, but the most consistent minimal criteria was recommended by the International Society for Cellular Therapy. Confirmation of the multipotent status of these isolated cells can be achieved by differentiation into the three basic mesodermal lineages in vitro.
Collapse
|
30
|
Shaikh MS, Zafar MS, Pisani F, Lone MA, Malik YR. Critical features of periodontal flaps with regard to blood clot stability: A review. J Oral Biosci 2021; 63:111-119. [PMID: 33684521 DOI: 10.1016/j.job.2021.02.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 02/22/2021] [Accepted: 02/24/2021] [Indexed: 12/12/2022]
Abstract
BACKGROUND Wound healing is a multifactorial procedure involving different cell types and biological mediators. The principles of wound healing are also applicable to periodontal tissues. The formation and stability of blood clots play a vital role in successful healing of wounds in periodontal tissues. The aim of the present review was to highlight the vital factors of periodontal flaps associated with blood clot stability. HIGHLIGHT The data on periodontal regeneration and wound healing have evolved greatly in light of several factors, including space for blood clots and blood clot stabilization. In periodontal osseous defects, the stability of blood clots seems critical to wound healing. If mechanical forces can be managed by wound stabilization, the gingival flap-tooth root interface may show connective tissue repair. However, compromised adhesion is susceptible to mechanical forces and can cause wound breakage and epithelialization. CONCLUSION The presence of a thick blood clot may hinder the plasmatic circulation between the recipient bed and graft during the initial stage of healing, which is critical in cases of mucogingival surgery. Root conditioning can also determine the healing consequence by enhancing blood clot adhesion.
Collapse
Affiliation(s)
- Muhammad Saad Shaikh
- Department of Oral Biology, Sindh Institute of Oral Health Sciences, Jinnah Sindh Medical University, Karachi, 75510, Pakistan
| | - Muhammad Sohail Zafar
- Department of Restorative Dentistry, College of Dentistry, Taibah University, Madina Munawwarra, 41311, Saudi Arabia; Department of Dental Materials, Islamic International Dental College, Riphah International University, Islamabad, 44000, Pakistan.
| | - Flavio Pisani
- College of Medicine and Dentistry, MClinDent in Periodontology, Birmingham, B4 6BN, UK
| | - Mohid Abrar Lone
- Department of Oral Pathology, Sindh Institute of Oral Health Sciences, Jinnah Sindh Medical University, Karachi, 75510, Pakistan
| | - Yasser Riaz Malik
- Department of Community Dentistry, Sir Syed College of Medical Sciences for Girls, Karachi, 74200, Pakistan; Department of Preventive Dentistry (Dental Public Health), College of Dentistry, University of Hail, Hail, Saudi Arabia
| |
Collapse
|
31
|
Amler AK, Dinkelborg PH, Schlauch D, Spinnen J, Stich S, Lauster R, Sittinger M, Nahles S, Heiland M, Kloke L, Rendenbach C, Beck-Broichsitter B, Dehne T. Comparison of the Translational Potential of Human Mesenchymal Progenitor Cells from Different Bone Entities for Autologous 3D Bioprinted Bone Grafts. Int J Mol Sci 2021; 22:E796. [PMID: 33466904 PMCID: PMC7830021 DOI: 10.3390/ijms22020796] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 12/28/2020] [Accepted: 01/11/2021] [Indexed: 02/08/2023] Open
Abstract
Reconstruction of segmental bone defects by autologous bone grafting is still the standard of care but presents challenges including anatomical availability and potential donor site morbidity. The process of 3D bioprinting, the application of 3D printing for direct fabrication of living tissue, opens new possibilities for highly personalized tissue implants, making it an appealing alternative to autologous bone grafts. One of the most crucial hurdles for the clinical application of 3D bioprinting is the choice of a suitable cell source, which should be minimally invasive, with high osteogenic potential, with fast, easy expansion. In this study, mesenchymal progenitor cells were isolated from clinically relevant human bone biopsy sites (explant cultures from alveolar bone, iliac crest and fibula; bone marrow aspirates; and periosteal bone shaving from the mastoid) and 3D bioprinted using projection-based stereolithography. Printed constructs were cultivated for 28 days and analyzed regarding their osteogenic potential by assessing viability, mineralization, and gene expression. While viability levels of all cell sources were comparable over the course of the cultivation, cells obtained by periosteal bone shaving showed higher mineralization of the print matrix, with gene expression data suggesting advanced osteogenic differentiation. These results indicate that periosteum-derived cells represent a highly promising cell source for translational bioprinting of bone tissue given their superior osteogenic potential as well as their minimally invasive obtainability.
Collapse
Affiliation(s)
- Anna-Klara Amler
- Department of Medical Biotechnology, Technische Universität Berlin, 13355 Berlin, Germany; (A.-K.A.); (D.S.); (R.L.)
- Cellbricks GmbH, 13355 Berlin, Germany;
| | - Patrick H. Dinkelborg
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt Universität zu Berlin, Department of Oral and Maxillofacial Surgery, and Berlin Institute of Health, 13353 Berlin, Germany; (S.N.); (M.H.); (C.R.); (B.B.-B.)
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt Universität zu Berlin, Department of Rheumatology, and Berlin Institute of Health, 10117 Berlin, Germany; (J.S.); (S.S.); (M.S.); (T.D.)
| | - Domenic Schlauch
- Department of Medical Biotechnology, Technische Universität Berlin, 13355 Berlin, Germany; (A.-K.A.); (D.S.); (R.L.)
- Cellbricks GmbH, 13355 Berlin, Germany;
| | - Jacob Spinnen
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt Universität zu Berlin, Department of Rheumatology, and Berlin Institute of Health, 10117 Berlin, Germany; (J.S.); (S.S.); (M.S.); (T.D.)
| | - Stefan Stich
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt Universität zu Berlin, Department of Rheumatology, and Berlin Institute of Health, 10117 Berlin, Germany; (J.S.); (S.S.); (M.S.); (T.D.)
| | - Roland Lauster
- Department of Medical Biotechnology, Technische Universität Berlin, 13355 Berlin, Germany; (A.-K.A.); (D.S.); (R.L.)
| | - Michael Sittinger
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt Universität zu Berlin, Department of Rheumatology, and Berlin Institute of Health, 10117 Berlin, Germany; (J.S.); (S.S.); (M.S.); (T.D.)
| | - Susanne Nahles
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt Universität zu Berlin, Department of Oral and Maxillofacial Surgery, and Berlin Institute of Health, 13353 Berlin, Germany; (S.N.); (M.H.); (C.R.); (B.B.-B.)
| | - Max Heiland
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt Universität zu Berlin, Department of Oral and Maxillofacial Surgery, and Berlin Institute of Health, 13353 Berlin, Germany; (S.N.); (M.H.); (C.R.); (B.B.-B.)
| | | | - Carsten Rendenbach
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt Universität zu Berlin, Department of Oral and Maxillofacial Surgery, and Berlin Institute of Health, 13353 Berlin, Germany; (S.N.); (M.H.); (C.R.); (B.B.-B.)
| | - Benedicta Beck-Broichsitter
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt Universität zu Berlin, Department of Oral and Maxillofacial Surgery, and Berlin Institute of Health, 13353 Berlin, Germany; (S.N.); (M.H.); (C.R.); (B.B.-B.)
| | - Tilo Dehne
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt Universität zu Berlin, Department of Rheumatology, and Berlin Institute of Health, 10117 Berlin, Germany; (J.S.); (S.S.); (M.S.); (T.D.)
| |
Collapse
|
32
|
Chen A, Tong YW, Chiu CH, Lei KF. Osteogenic Effect of Rabbit Periosteum-Derived Precursor Cells Co-Induced by Electric Stimulation and Adipose-Derived Stem Cells in a 3D Co-Culture System. IEEE OPEN JOURNAL OF NANOTECHNOLOGY 2021; 2:153-160. [DOI: 10.1109/ojnano.2021.3131653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/23/2025]
|
33
|
Naujokat H, Loger K, Schulz J, Açil Y, Wiltfang J. Bone tissue engineering in the greater omentum with computer-aided design/computer-aided manufacturing scaffolds is enhanced by a periosteum transplant. Regen Med 2020; 15:2297-2309. [PMID: 33355523 DOI: 10.2217/rme-2020-0115] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Aim: This study aimed to evaluate two different vascularized bone flap scaffolds and the impact of two barrier membranes for the reconstruction of critical-size bone defects. Materials & methods: 3D-printed scaffolds of biodegradable calcium phosphate and bioinert titanium were loaded with rhBMP-2 bone marrow aspirate, wrapped by a collagen membrane or a periosteum transplant and implanted into the greater omentum of miniature pigs. Results: Histological evaluation demonstrated significant bone formation within the first 8 weeks in both scaffolds. The periosteum transplant led to enhanced bone formation and a homogenous distribution in the scaffolds. The omentum tissue grew out a robust vascular supply. Conclusion: Endocultivation using 3D-printed scaffolds in the greater omentum is a very promising approach in defect-specific bone tissue regeneration.
Collapse
Affiliation(s)
- Hendrik Naujokat
- Department of Oral & Maxillofacial Surgery, University Hospital Schleswig-Holstein, Campus Kiel, Arnold-Heller-Straße 3, 24105 Kiel, Germany
| | - Klaas Loger
- Department of Oral & Maxillofacial Surgery, University Hospital Schleswig-Holstein, Campus Kiel, Arnold-Heller-Straße 3, 24105 Kiel, Germany
| | - Juliane Schulz
- Department of Oral & Maxillofacial Surgery, University Hospital Schleswig-Holstein, Campus Kiel, Arnold-Heller-Straße 3, 24105 Kiel, Germany
| | - Yahya Açil
- Department of Oral & Maxillofacial Surgery, University Hospital Schleswig-Holstein, Campus Kiel, Arnold-Heller-Straße 3, 24105 Kiel, Germany
| | - Jörg Wiltfang
- Department of Oral & Maxillofacial Surgery, University Hospital Schleswig-Holstein, Campus Kiel, Arnold-Heller-Straße 3, 24105 Kiel, Germany
| |
Collapse
|
34
|
Jin Y, Sun X, Pei F, Zhao Z, Mao J. Wnt16 signaling promotes osteoblast differentiation of periosteal derived cells in vitro and in vivo. PeerJ 2020; 8:e10374. [PMID: 33282557 PMCID: PMC7694570 DOI: 10.7717/peerj.10374] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 10/26/2020] [Indexed: 02/05/2023] Open
Abstract
Background Periosteum plays critical roles in de novo bone formation and fracture repair. Wnt16 has been regarded as a key regulator in periosteum bone formation. However, the role of Wnt16 in periosteum derived cells (PDCs) osteogenic differentiation remains unclear. The study goal is to uncover whether and how Wnt16 acts on the osteogenesis of PDCs. Methods We detected the variation of Wnt16 mRNA expression in PDCs, which were isolated from mouse femur and identified by flow cytometry, cultured in osteogenic medium for 14 days, then knocked down and over-expressed Wnt16 in PDCs to analysis its effects in osteogenesis. Further, we seeded PDCs (Wnt16 over-expressed/vector) in β-tricalcium phosphate cubes, and transplanted this complex into a critical size calvarial defect. Lastly, we used immunofluorescence, Topflash and NFAT luciferase reporter assay to study the possible downstream signaling pathway of Wnt16. Results Wnt16 mRNA expression showed an increasing trend in PDCs under osteogenic induction for 14 days. Wnt16 shRNA reduced mRNA expression of Runx2, collage type I (Col-1) and osteocalcin (OCN) after 7 days of osteogenic induction, as well as alizarin red staining intensity after 21days. Wnt16 also increased the mRNA expression of Runx2 and OCN and the protein production of Runx2 and Col-1 after 2 days of osteogenic stimulation. In the orthotopic transplantation assay, more bone volume, trabecula number and less trabecula space were found in Wnt16 over-expressed group. Besides, in the newly formed tissue Brdu positive area was smaller and Col-1 was larger in Wnt16 over-expressed group compared to the control group. Finally, Wnt16 upregulated CTNNB1/β-catenin expression and its nuclear translocation in PDCs, also increased Topflash reporter luciferase activity. By contrast, Wnt16 failed to increase NFAT reporter luciferase activity. Conclusion Together, Wnt16 plays a positive role in regulating PDCs osteogenesis, and Wnt16 may have a potential use in improving bone regeneration.
Collapse
Affiliation(s)
- Ying Jin
- Department of Orthodontics, State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, West China School of Stomatology, Sichuan University, Chengdu, China
| | - Xiaoyan Sun
- Stomatological Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Fang Pei
- Department of Orthodontics, State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, West China School of Stomatology, Sichuan University, Chengdu, China
| | - Zhihe Zhao
- Department of Orthodontics, State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, West China School of Stomatology, Sichuan University, Chengdu, China
| | - Jeremy Mao
- Columbia University, Center for Craniofacial Regeneration, New York, NY, United States of America
| |
Collapse
|
35
|
Watanabe T, Takabatake K, Tsujigiwa H, Watanabe S, Nakagiri R, Nakano K, Nagatsuka H, Kimata Y. Effect of Honeycomb β-TCP Geometrical Structure on Bone Tissue Regeneration in Skull Defect. MATERIALS (BASEL, SWITZERLAND) 2020; 13:E4761. [PMID: 33113818 PMCID: PMC7663559 DOI: 10.3390/ma13214761] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 10/23/2020] [Accepted: 10/23/2020] [Indexed: 11/30/2022]
Abstract
The effect of the geometric structure of artificial biomaterials on skull regeneration remains unclear. In a previous study, we succeeded in developing honeycomb β-tricalcium phosphate (β-TCP), which has through-and-through holes and is able to provide the optimum bone microenvironment for bone tissue regeneration. We demonstrated that β-TCP with 300-μm hole diameters induced vigorous bone formation. In the present study, we investigated how differences in hole directions of honeycomb β-TCP (horizontal or vertical holes) influence bone tissue regeneration in skull defects. Honeycomb β-TCP with vertical and horizontal holes was loaded with BMP-2 using Matrigel and Collagen gel as carriers, and transplanted into skull bone defect model rats. The results showed that in each four groups (Collagen alone group, Matrigel alone group, Collagen + BMP group and Matrigel + BMP-2), vigorous bone formation was observed on the vertical β-TCP compared with horizontal β-TCP. The osteogenic area was larger in the Matrigel groups (with and without BMP-2) than in the Collagen group (with and without BMP-2) in both vertical β-TCP and horizontal β-TCP. However, when BMP-2 was added, the bone formation area was not significantly different between the Collagen group and the Matrigel group in the vertical β-TCP. Histological finding showed that, in vertical honeycomb β-TCP, new bone formation extended to the upper part of the holes and was observed from the dura side to the periosteum side as added to the inner walls of the holes. Therefore, we can control efficient bone formation by creating a bone microenvironment provided by vertical honeycomb β-TCP. Vertical honeycomb β-TCP has the potential to be an excellent biomaterial for bone tissue regeneration in skull defects and is expected to have clinical applications.
Collapse
Affiliation(s)
- Toshiyuki Watanabe
- Department of Plastic and Reconstructive Surgery, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 7008525, Japan; (T.W.); (S.W.); (R.N.); (Y.K.)
| | - Kiyofumi Takabatake
- Department of Oral Pathology and Medicine, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 7008525, Japan; (K.N.); (H.N.)
| | - Hidetsugu Tsujigiwa
- Department of Life Science, Faculty of Science, Okayama University Science, Okayama 7000005, Japan;
| | - Satoko Watanabe
- Department of Plastic and Reconstructive Surgery, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 7008525, Japan; (T.W.); (S.W.); (R.N.); (Y.K.)
| | - Ryoko Nakagiri
- Department of Plastic and Reconstructive Surgery, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 7008525, Japan; (T.W.); (S.W.); (R.N.); (Y.K.)
| | - Keisuke Nakano
- Department of Oral Pathology and Medicine, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 7008525, Japan; (K.N.); (H.N.)
| | - Hitoshi Nagatsuka
- Department of Oral Pathology and Medicine, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 7008525, Japan; (K.N.); (H.N.)
| | - Yoshihiro Kimata
- Department of Plastic and Reconstructive Surgery, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 7008525, Japan; (T.W.); (S.W.); (R.N.); (Y.K.)
| |
Collapse
|
36
|
Borciani G, Montalbano G, Baldini N, Cerqueni G, Vitale-Brovarone C, Ciapetti G. Co-culture systems of osteoblasts and osteoclasts: Simulating in vitro bone remodeling in regenerative approaches. Acta Biomater 2020; 108:22-45. [PMID: 32251782 DOI: 10.1016/j.actbio.2020.03.043] [Citation(s) in RCA: 109] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Revised: 03/20/2020] [Accepted: 03/30/2020] [Indexed: 02/08/2023]
Abstract
Bone is an extremely dynamic tissue, undergoing continuous remodeling for its whole lifetime, but its regeneration or augmentation due to bone loss or defects are not always easy to obtain. Bone tissue engineering (BTE) is a promising approach, and its success often relies on a "smart" scaffold, as a support to host and guide bone formation through bone cell precursors. Bone homeostasis is maintained by osteoblasts (OBs) and osteoclasts (OCs) within the basic multicellular unit, in a consecutive cycle of resorption and formation. Therefore, a functional scaffold should allow the best possible OB/OC cooperation for bone remodeling, as happens within the bone extracellular matrix in the body. In the present work OB/OC co-culture models, with and without scaffolds, are reviewed. These experimental systems are intended for different targets, including bone remodeling simulation, drug testing and the assessment of biomaterials and 3D scaffolds for BTE. As a consequence, several parameters, such as cell type, cell ratio, culture medium and inducers, culture times and setpoints, assay methods, etc. vary greatly. This review identifies and systematically reports the in vitro methods explored up to now, which, as they allow cellular communication, more closely resemble bone remodeling and/or the regeneration process in the framework of BTE. STATEMENT OF SIGNIFICANCE: Bone is a dynamic tissue under continuous remodeling, but spontaneous healing may fail in the case of excessive bone loss which often requires valid alternatives to conventional treatments to restore bone integrity, like bone tissue engineering (BTE). Pre-clinical evaluation of scaffolds for BTE requires in vitro testing where co-cultures combining innovative materials with osteoblasts (OBs) and osteoclasts (OCs) closely mimic the in vivo repair process. This review considers the direct and indirect OB/OC co-cultures relevant to BTE, from the early mouse-cell models to the recent bone regenerative systems. The co-culture modeling of bone microenvironment provides reliable information on bone cell cross-talk. Starting from improved knowledge on bone remodeling, bone disease mechanisms may be understood and new BTE solutions are designed.
Collapse
|
37
|
Umrath F, Weber M, Reinert S, Wendel HP, Avci-Adali M, Alexander D. iPSC-Derived MSCs Versus Originating Jaw Periosteal Cells: Comparison of Resulting Phenotype and Stem Cell Potential. Int J Mol Sci 2020; 21:ijms21020587. [PMID: 31963278 PMCID: PMC7013802 DOI: 10.3390/ijms21020587] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 01/04/2020] [Accepted: 01/14/2020] [Indexed: 02/07/2023] Open
Abstract
Induced pluripotent stem cell-derived mesenchymal stem cell-like cells (iMSCs) are considered to be a promising source of progenitor cells for approaches in the field of bone regeneration. In a previous study, we described the generation of footprint-free induced pluripotent stem cells (iPSCs) from human jaw periosteal cells (JPCs) by transfection of a self-replicating RNA (srRNA) and subsequent differentiation into functional osteogenic progenitor cells. In order to facilitate the prospective transfer into clinical practice, xeno-free reprogramming and differentiation methods were established. In this study, we compared the properties and stem cell potential of the iMSCs produced from JPC-derived iPSCs with the parental primary JPCs they were generated from. Our results demonstrated, on the one hand, a comparable differentiation potential of iMSCs and JPCs. Additionally, iMSCs showed significantly longer telomere lengths compared to JPCs indicating rejuvenation of the cells during reprogramming. On the other hand, proliferation, mitochondrial activity, and senescence-associated beta-galactosidase (SA-β-gal) activity indicated early senescence of iMSCs. These data demonstrate the requirement of further optimization strategies to improve mesenchymal development of JPC-derived iPSCs in order to take advantage of the best features of reprogrammed and rejuvenated cells.
Collapse
Affiliation(s)
- Felix Umrath
- Department of Oral and Maxillofacial Surgery, University Hospital Tübingen, 72076 Tübingen, Germany; (F.U.); (S.R.)
| | - Marbod Weber
- Department of Thoracic and Cardiovascular Surgery, University Hospital Tübingen, 72076 Tübingen, Germany; (M.W.); (H.-P.W.); (M.A.-A.)
| | - Siegmar Reinert
- Department of Oral and Maxillofacial Surgery, University Hospital Tübingen, 72076 Tübingen, Germany; (F.U.); (S.R.)
| | - Hans-Peter Wendel
- Department of Thoracic and Cardiovascular Surgery, University Hospital Tübingen, 72076 Tübingen, Germany; (M.W.); (H.-P.W.); (M.A.-A.)
| | - Meltem Avci-Adali
- Department of Thoracic and Cardiovascular Surgery, University Hospital Tübingen, 72076 Tübingen, Germany; (M.W.); (H.-P.W.); (M.A.-A.)
| | - Dorothea Alexander
- Department of Oral and Maxillofacial Surgery, University Hospital Tübingen, 72076 Tübingen, Germany; (F.U.); (S.R.)
- Correspondence: ; Tel.: +49-7071-29-82418
| |
Collapse
|
38
|
Shahlaei M, Asl SM, Saeidifar M. Nanotechnology in gene delivery for neural regenerative medicine. NEURAL REGENERATIVE NANOMEDICINE 2020:123-157. [DOI: 10.1016/b978-0-12-820223-4.00005-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
39
|
Lee AR, Moon DK, Siregar A, Moon SY, Jeon RH, Son YB, Kim BG, Hah YS, Hwang SC, Byun JH, Woo DK. Involvement of mitochondrial biogenesis during the differentiation of human periosteum-derived mesenchymal stem cells into adipocytes, chondrocytes and osteocytes. Arch Pharm Res 2019; 42:1052-1062. [PMID: 31802425 DOI: 10.1007/s12272-019-01198-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 11/27/2019] [Indexed: 02/07/2023]
Abstract
Due to a rapidly expanding aging population, the incidence of age-related or degenerative diseases has increased, and efforts to handle the issue with regenerative medicine via adult stem cells have become more important. And it is now clear that the mitochondrial energy metabolism is important for stem cell differentiation. When stem cells commit to differentiate, glycolytic metabolism is being shifted to mitochondrial oxidative phosphorylation (OXPHOS) to meet an increased cellular energy demand required for differentiated cells. However, the nature of cellular metabolisms during the differentiation process of periosteum-derived mesenchymal stem cells (POMSC) is still unclear. In the present study, we investigated mitochondrial biogenesis during the adipogenic, chondrogenic, and osteogenic differentiation of POMSCs. Both mitochondrial DNA (mtDNA) contents and mitochondrial proteins (VDAC and mitochondrial OXPHOS complex subunits) were increased during all of these mesenchymal lineage differentiations of POMSCs. Interestingly, glycolytic metabolism is reduced as POMSCs undergo osteogenic differentiation. Furthermore, reducing mtDNA contents by ethidium bromide treatments prevents osteogenic differentiation of POMSCs. In conclusion, these results indicate that mitochondrial biogenesis and OXPHOS metabolism play important roles in the differentiation of POMCS and suggest that pharmaceutical modulation of mitochondrial biogenesis and/or function can be a novel regulation for POMSC differentiation and regenerative medicine.
Collapse
Affiliation(s)
- A Ram Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Gyeongsang National University, Jinju, Republic of Korea
| | - Dong Kyu Moon
- Department of Orthopedic Surgery and Institute of Health Sciences, School of Medicine and Hospital, Gyeongsang National University, Jinju, Republic of Korea
| | - Adrian Siregar
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Gyeongsang National University, Jinju, Republic of Korea
| | - Sun Young Moon
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Gyeongsang National University, Jinju, Republic of Korea
| | - Ryoung-Hoon Jeon
- Department of Theriogenology and Biotechnology, College of Veterinary Medicine, Gyeongsang National University, Jinju, Republic of Korea
| | - Young-Bum Son
- Department of Theriogenology and Biotechnology, College of Veterinary Medicine, Gyeongsang National University, Jinju, Republic of Korea
| | - Bo Gyu Kim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Gyeongsang National University, Jinju, Republic of Korea
| | - Young-Sool Hah
- Clinical Research Institute of Gyeongsang, National University Hospital, Jinju, Republic of Korea
| | - Sun-Chul Hwang
- Department of Orthopedic Surgery and Institute of Health Sciences, School of Medicine and Hospital, Gyeongsang National University, Jinju, Republic of Korea
| | - June-Ho Byun
- Department of Oral and Maxillofacial Surgery and Institute of Health Sciences, School of Medicine and Hospital, Gyeongsang National University, Jinju, Republic of Korea.
| | - Dong Kyun Woo
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Gyeongsang National University, Jinju, Republic of Korea.
| |
Collapse
|
40
|
Zhao Z, Fan C, Chen F, Sun Y, Xia Y, Ji A, Wang DA. Progress in Articular Cartilage Tissue Engineering: A Review on Therapeutic Cells and Macromolecular Scaffolds. Macromol Biosci 2019; 20:e1900278. [PMID: 31800166 DOI: 10.1002/mabi.201900278] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2019] [Revised: 09/19/2019] [Indexed: 12/19/2022]
Abstract
Repair and regeneration of articular cartilage lesions have always been a major challenge in the medical field due to its peculiar structure (e.g., sparsely distributed chondrocytes, no blood supply, no nerves). Articular cartilage tissue engineering is considered as one promising strategy to achieve reconstruction of cartilage. With this perspective, the articular cartilage tissue engineering has been widely studied. Here, the recent progress of articular cartilage tissue engineering is reviewed. The ad hoc therapeutic cells and growth factors for cartilage regeneration are summarized and discussed. Various types of bio/macromolecular scaffolds together with their pros and cons are also reviewed and elaborated.
Collapse
Affiliation(s)
- Zhongyi Zhao
- Department of Traumatic Surgery, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Changjiang Fan
- Department of Human Anatomy, Histology and Embryology, College of Medicine, Qingdao University, Qingdao, 266021, China.,Institute for Translational Medicine, College of Medicine, Qingdao University, Qingdao, P. R. China
| | - Feng Chen
- Department of Traumatic Surgery, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yutai Sun
- School of Information Engineering, Shandong Vocational College of Science & Technology, Weifang, 261053, P. R. China
| | - Yujun Xia
- Department of Human Anatomy, Histology and Embryology, College of Medicine, Qingdao University, Qingdao, 266021, China
| | - Aiyu Ji
- Department of Traumatic Surgery, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Dong-An Wang
- Department of Biomedical Engineering, City University of Hong Kong, Kowloon, 999077, Hong Kong SAR
| |
Collapse
|
41
|
Quality Analysis of Minerals Formed by Jaw Periosteal Cells under Different Culture Conditions. Int J Mol Sci 2019; 20:ijms20174193. [PMID: 31461878 PMCID: PMC6747376 DOI: 10.3390/ijms20174193] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 08/16/2019] [Accepted: 08/22/2019] [Indexed: 02/06/2023] Open
Abstract
Previously, we detected a higher degree of mineralization in fetal calf serum (FCS) compared to serum-free cultured jaw periosteum derived osteoprogenitor cells (JPCs). By Raman spectroscopy, we detected an earlier formation of mineralized extracellular matrix (ECM) of higher quality under serum-free media conditions. However, mineralization potential remained too low. In the present study, we aimed to investigate the biochemical composition and subsequent biomechanical properties of the JPC-formed ECM and minerals under human platelet lysate (hPL) and FCS supplementation. JPCs were isolated (n = 4 donors) and expanded under FCS conditions and used in passage five for osteogenic induction under both, FCS and hPL media supplementation. Raman spectroscopy and Alizarin Red/von Kossa staining were employed for biochemical composition analyses and for visualization and quantification of mineralization. Osteocalcin gene expression was analyzed by quantitative PCR. Biomechanical properties were assessed by using atomic force microscopy (AFM). Raman spectroscopic measurements showed significantly higher (p < 0.001) phosphate to protein ratios and in the tendency, lower carbonate to phosphate ratios in osteogenically induced JPCs under hPL in comparison to FCS culturing. Furthermore, higher crystal sizes were detected under hPL culturing of the cells. With respect to the ECM, significantly higher ratios of the precursor protein proline to hydroxyproline were detected in hPL-cultured JPC monolayers (p < 0.001). Additionally, significantly higher levels (p < 0.001) of collagen cross-linking were calculated, indicating a higher degree of collagen maturation in hPL-cultured JPCs. By atomic force microscopy, a significant increase in ECM stiffness (p < 0.001) of FCS cultured JPC monolayers was observed. The reverse effect was measured for the JPC formed precipitates/minerals. Under hPL supplementation, JPCs formed minerals of significantly higher stiffness (p < 0.001) when compared to the FCS setting. This study demonstrates that hPL culturing of JPCs leads to the formation of an anorganic material of superior quality in terms of biochemical composition and mechanical properties.
Collapse
|
42
|
Park GH, Shin HS, Choi ES, Yoon BS, Choi MH, Lee SJ, Lee KE, Lee JS, Hong JM. Cranial burr hole with erythropoietin administration induces reverse arteriogenesis from the enriched extracranium. Neurobiol Dis 2019; 132:104538. [PMID: 31344491 DOI: 10.1016/j.nbd.2019.104538] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 06/07/2019] [Accepted: 07/19/2019] [Indexed: 01/21/2023] Open
Abstract
It is challenging to revitalize ischemic penumbra after an acute stroke with intracranial perfusion insufficiency. To evaluate whether cranial burr hole and erythropoietin (EPO) generate effective revascularization, we investigated the efficacy of the augmentation method for reverse arteriogenesis from the healthy extracranial milieu. An intracranial perfusion insufficiency was created through bilateral internal carotid artery ligation (bICAL) in Sprague-Dawley rats. We administered recombinant human EPO (5000 U/kg) or saline intraperitoneally for 3 days after bICAL. Mechanical barrier disruption (MBD) was performed through a cranial burr hole with small dural cracks in the right hemisphere. The ipsilateral hemisphere with MBD grossly showed vascular networks between the extra- and intra-cranial spaces 2 weeks after the MBD procedure. It also showed significantly increased vessels in the intracranial vasculature adjacent to the MBD region (p = 0.0006). The levels of pro-angiogenic and inflammatory factors with prominent markers of vessel permeability were also significantly increased (MBD-only vs. control; Tnf-α, p = 0.0007; Vegf, p = 0.0206). In the EPO-administered group, such elevations in inflammation were significantly mitigated (combined vs. MBD-only; Tnf-α, p = 0.0008). The ipsilateral hemisphere with MBD-EPO (vs. MBD-only) showed significantly increased vessels (RECA-1, p = 0.0182) and their maturation (RECA-1/α-SMA, p = 0.0046), with upregulation of tumor growth factor-β1 (Tgf-β1, p = 0.037) and matrix metalloproteinase-2 (Mmp-2, p = 0.0488). These findings were completely blocked by minocycline (MIC) administration during in vivo (Tgf-β1, p = 0.0009; Mmp-2, p < 0.0001) and in vitro experiments (tube formation, p < 0.0001). Our data suggest that the MBD procedure (for angiogenic routes) and EPO administration (for an arteriogenic booster) are complimentary and can facilitate successfully "reverse arteriogenesis" in subjects with intracranial perfusion insufficiency.
Collapse
Affiliation(s)
- Geun Hwa Park
- Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon, South Korea
| | - Hee Sun Shin
- Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon, South Korea
| | - Eun Sil Choi
- Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon, South Korea
| | - Bok Seon Yoon
- Department of Neurology, Ajou University School of Medicine, Ajou University Medical Center, Suwon, South Korea
| | - Mun Hee Choi
- Department of Neurology, Ajou University School of Medicine, Ajou University Medical Center, Suwon, South Korea
| | - Seong-Joon Lee
- Department of Neurology, Ajou University School of Medicine, Ajou University Medical Center, Suwon, South Korea
| | - Kyung-Eon Lee
- Department of Life and Nanopharmaceutical Sciences, Kyung Hee University School of Pharmacy, Seoul, South Korea
| | - Jin Soo Lee
- Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon, South Korea; Department of Neurology, Ajou University School of Medicine, Ajou University Medical Center, Suwon, South Korea
| | - Ji Man Hong
- Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon, South Korea; Department of Neurology, Ajou University School of Medicine, Ajou University Medical Center, Suwon, South Korea.
| |
Collapse
|
43
|
Generation of iPSCs from Jaw Periosteal Cells Using Self-Replicating RNA. Int J Mol Sci 2019; 20:ijms20071648. [PMID: 30987077 PMCID: PMC6480539 DOI: 10.3390/ijms20071648] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Revised: 03/30/2019] [Accepted: 03/30/2019] [Indexed: 12/16/2022] Open
Abstract
Jaw periosteal cells (JPCs) represent a suitable stem cell source for bone tissue engineering (BTE) applications. However, challenges associated with limited cell numbers, stressful cell sorting, or the occurrence of cell senescence during in vitro passaging and the associated insufficient osteogenic potential in vitro of JPCs and other mesenchymal stem/stromal cells (MSCs) are main hurdles and still need to be solved. In this study, for the first time, induced pluripotent stem cells (iPSCs) were generated from human JPCs to open up a new source of stem cells for BTE. For this purpose, a non-integrating self-replicating RNA (srRNA) encoding reprogramming factors and green fluorescent protein (GFP) as a reporter was used to obtain JPC-iPSCs with a feeder- and xeno-free reprogramming protocol to meet the highest safety standards for future clinical applications. Furthermore, to analyze the potential of these iPSCs as a source of osteogenic progenitor cells, JPC-iPSCs were differentiated into iPSC-derived mesenchymal stem/stromal like cells (iMSCs) and further differentiated to the osteogenic lineage under xeno-free conditions. The produced iMSCs displayed MSC marker expression and morphology as well as strong mineralization during osteogenic differentiation.
Collapse
|
44
|
Caddeo S, Mattioli-Belmonte M, Cassino C, Barbani N, Dicarlo M, Gentile P, Baino F, Sartori S, Vitale-Brovarone C, Ciardelli G. Newly-designed collagen/polyurethane bioartificial blend as coating on bioactive glass-ceramics for bone tissue engineering applications. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2019; 96:218-233. [DOI: 10.1016/j.msec.2018.11.012] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 11/02/2018] [Accepted: 11/07/2018] [Indexed: 10/27/2022]
|
45
|
Naujokat H, Lipp M, Açil Y, Wieker H, Birkenfeld F, Sengebusch A, Böhrnsen F, Wiltfang J. Bone tissue engineering in the greater omentum is enhanced by a periosteal transplant in a miniature pig model. Regen Med 2019; 14:127-138. [DOI: 10.2217/rme-2018-0031] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Aim: Reconstruction of bone defects with autologous grafts has certain disadvantages. The aim of this study is to introduce a new type of living bioreactor for engineering of bone flaps and to evaluate the effect of different barrier membranes. Materials & methods: Scaffolds loaded with bone morphogenetic proteins and bone marrow aspirate wrapped with either a collagen membrane or a periosteal flap were implanted in the greater omentum of miniature pigs. Results: Both histological and radiographic evaluation showed proven bone formation and increased density after 8 and 16 weeks, with an enhanced effect of the periosteal transplant. Conclusion: The greater omentum is a suitable bioreactor for bone tissue engineering. Endocultivation is both an innovative and promising approach in regenerative medicine.
Collapse
Affiliation(s)
- Hendrik Naujokat
- Department of Oral & Maxillofacial Surgery, University Hospital of Schleswig-Holstein, Campus Kiel, Arnold-Heller-Straße 3, 24105 Kiel, Germany
| | - Maximilian Lipp
- Department of Oral & Maxillofacial Surgery, University Hospital of Schleswig-Holstein, Campus Kiel, Arnold-Heller-Straße 3, 24105 Kiel, Germany
| | - Yahya Açil
- Department of Oral & Maxillofacial Surgery, University Hospital of Schleswig-Holstein, Campus Kiel, Arnold-Heller-Straße 3, 24105 Kiel, Germany
| | - Henning Wieker
- Department of Oral & Maxillofacial Surgery, University Hospital of Schleswig-Holstein, Campus Kiel, Arnold-Heller-Straße 3, 24105 Kiel, Germany
| | - Falk Birkenfeld
- Department of Oral & Maxillofacial Surgery, University Hospital of Schleswig-Holstein, Campus Kiel, Arnold-Heller-Straße 3, 24105 Kiel, Germany
| | - Andre Sengebusch
- Department of Oral & Maxillofacial Surgery, University Hospital of Schleswig-Holstein, Campus Kiel, Arnold-Heller-Straße 3, 24105 Kiel, Germany
| | - Florian Böhrnsen
- Department of Oral & Maxillofacial Surgery, University Hospital of Göttingen, Robert-Koch-Straße 40, 37099 Göttingen, Germany
| | - Jörg Wiltfang
- Department of Oral & Maxillofacial Surgery, University Hospital of Schleswig-Holstein, Campus Kiel, Arnold-Heller-Straße 3, 24105 Kiel, Germany
| |
Collapse
|
46
|
Wang X, Matthews BG, Yu J, Novak S, Grcevic D, Sanjay A, Kalajzic I. PDGF Modulates BMP2-Induced Osteogenesis in Periosteal Progenitor Cells. JBMR Plus 2019; 3:e10127. [PMID: 31131345 PMCID: PMC6524680 DOI: 10.1002/jbm4.10127] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 09/23/2018] [Accepted: 10/03/2018] [Indexed: 12/15/2022] Open
Abstract
BMPs are used in various clinical applications to promote bone formation. The limited success of the BMPs in clinical settings and supraphysiological doses required for their effects prompted us to evaluate the influence of other signaling molecules, specifically platelet‐derived growth factor (PDGF) on BMP2‐induced osteogenesis. Periosteal cells make a major contribution to fracture healing. We detected broad expression of PDGF receptor beta (PDGFRβ) within the intact periosteum and healing callus during fracture repair. In vitro, periosteum‐derived progenitor cells were highly responsive to PDGF as demonstrated by increased proliferation and decreased apoptosis. However, PDGF blocked BMP2‐induced osteogenesis by inhibiting the canonical BMP2/Smad pathway and downstream target gene expression. This effect is mediated via PDGFRβ and involves ERK1/2 MAPK and PI3K/AKT signaling pathways. Therapeutic targeting of the PDGFRβ pathway in periosteum‐mediated bone repair might have profound implications in the treatment of bone disease in the future. © 2018 The Authors JBMR Plus is published by Wiley Periodicals, Inc. on behalf of the American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Xi Wang
- Department of Reconstructive Sciences UConn Health Farmington CT USA
| | - Brya G Matthews
- Department of Reconstructive Sciences UConn Health Farmington CT USA.,Department of Molecular Medicine and Pathology University of Auckland Auckland New Zealand
| | - Jungeun Yu
- Department of Orthopedic Surgery UConn Health Farmington CT USA
| | - Sanja Novak
- Department of Reconstructive Sciences UConn Health Farmington CT USA
| | - Danka Grcevic
- Department of Physiology and Immunology School of Medicine University of Zagreb Zagreb Croatia
| | - Archana Sanjay
- Department of Orthopedic Surgery UConn Health Farmington CT USA
| | - Ivo Kalajzic
- Department of Reconstructive Sciences UConn Health Farmington CT USA
| |
Collapse
|
47
|
Bone Tissue Engineering Using Human Cells: A Comprehensive Review on Recent Trends, Current Prospects, and Recommendations. APPLIED SCIENCES-BASEL 2019. [DOI: 10.3390/app9010174] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The use of proper cells for bone tissue engineering remains a major challenge worldwide. Cells play a pivotal role in the repair and regeneration of the bone tissue in vitro and in vivo. Currently, a large number of differentiated (somatic) and undifferentiated (stem) cells have been used for bone reconstruction alone or in combination with different biomaterials and constructs (e.g., scaffolds). Although the results of the cell transplantation without any supporting or adjuvant material have been very effective with regard to bone healing. Recent advances in bone scaffolding are now becoming new players affecting the osteogenic potential of cells. In the present study, we have critically reviewed all the currently used cell sources for bone reconstruction and discussed the new horizons that are opening up in the context of cell-based bone tissue engineering strategies.
Collapse
|
48
|
Duchamp de Lageneste O, Colnot C. Periostin in Bone Regeneration. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1132:49-61. [PMID: 31037624 DOI: 10.1007/978-981-13-6657-4_6] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Bone regeneration is an efficient regenerative process depending on the recruitment and activation of skeletal stem cells that allow cartilage and bone formation leading to fracture consolidation. Periosteum, the tissue located at the outer surface of bone is now recognized as an essential player in the bone repair process and contains skeletal stem cells with high regenerative potential. The matrix composition of the periosteum defines its roles in bone growth, in cortical bone modeling and remodeling in response to mechanical strain, and in bone repair. Periostin is a key extracellular matrix component of the periosteum involved in periosteum functions. In this chapter, we summarize the current knowledge on the bone regeneration process, the role of the periosteum and skeletal stem cells, and Periostin functions in this context. The matricellular protein Periostin has several roles through all stages of bone repair: in the early days of repair during the initial activation of stem cells within periosteum, in the active phase of cartilage and bone deposition in the facture callus, and in the final phase of bone bridging and reconstitution of the stem cell pool within periosteum.
Collapse
Affiliation(s)
| | - Céline Colnot
- INSERM UMR1163, Imagine Institute, Paris Descartes University, Paris, France.
| |
Collapse
|
49
|
Effects of Jaw Periosteal Cells on Dendritic Cell Maturation. J Clin Med 2018; 7:jcm7100312. [PMID: 30274241 PMCID: PMC6210277 DOI: 10.3390/jcm7100312] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Revised: 09/25/2018] [Accepted: 09/26/2018] [Indexed: 12/16/2022] Open
Abstract
Clinical application of tissue engineering products requires the exclusion of immune responses after implantation. We used jaw periosteal cells (JPCs) as a suitable stem cell source and analyzed herein the effects of JPCs on dendritic cell maturation after co-culturing of both cell types. Peripheral blood mononuclear cells (PBMCs) were differentiated to dendritic cells (DCs) by the addition of differentiation cocktails for 7 days in co-culture with undifferentiated and osteogenically induced JPCs. The effects of JPCs on DC maturation were analyzed at the beginning (day 7), in the middle (day 14), and at the end (day 21) of the osteogenesis process. We detected significantly lower DC numbers after co-culturing with JPCs that have previously been left untreated or osteogenically differentiated for 7, 14, and 21 days. Using gene expression analyses, significantly lower IL-12p35 and -p40 and pro-inflammatory cytokine (IFN-γ and TNF-α) levels were detected, whereas IL-8 mRNA levels were significantly higher in DCs. Furthermore, osteogenic media conditions enhanced significantly IL-10 gene expression. We concluded that undifferentiated and osteogenically differentiated JPCs had an overall inhibiting influence on dendritic cell maturation. Further studies should clarify the underlaying mechanism in depth.
Collapse
|
50
|
Akiyama M. FBXW2 localizes with osteocalcin in bovine periosteum on culture dishes as visualized by double immunostaining. Heliyon 2018; 4:e00782. [PMID: 30229138 PMCID: PMC6141272 DOI: 10.1016/j.heliyon.2018.e00782] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 08/23/2018] [Accepted: 09/07/2018] [Indexed: 12/24/2022] Open
Abstract
Osteocalcin (OC) is a well-known protein related to bone, however, the role of F-box and WD-40 domain-containing protein 2 (FBXW2) in bone remains unclear. In 2016, the presence of FBXW2 in bovine periosteum was reported. In this study, double immunostaining was used to investigate the relationship between OC and FBXW2. FBXW2 showed tubular structures, and OC showed a similar localization pattern as FBXW2. Double immunostaining findings suggested that FBXW2 tubes were coated with OC. To the author's knowledge, this is the first study to reveal the interaction between OC and FBXW2.
Collapse
Affiliation(s)
- Mari Akiyama
- Department of Biomaterials, Osaka Dental University, 8-1, Kuzuhahanozono-cho, Hirakata-shi, Osaka 573-1121, Japan
| |
Collapse
|