1
|
Fiehn LA, Kunisch E, Saur M, Arango-Ospina M, Merle C, Hagmann S, Stiller A, Hupa L, Kaňková H, Galusková D, Renkawitz T, Boccaccini AR, Westhauser F. A comparative in vitro and in vivo analysis of the impact of copper substitution on the cytocompatibility, osteogenic, and angiogenic properties of a borosilicate bioactive glass. J Biomed Mater Res A 2024; 112:1740-1759. [PMID: 38623001 DOI: 10.1002/jbm.a.37721] [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: 01/21/2024] [Revised: 03/10/2024] [Accepted: 04/01/2024] [Indexed: 04/17/2024]
Abstract
The 0106-B1-bioactive glass (BG) composition (in wt %: 37.5 SiO2, 22.6 CaO, 5.9 Na2O, 4.0 P2O5, 12.0 K2O, 5.5 MgO, and 12.5 B2O3) has demonstrated favorable processing properties and promising bone regeneration potential. The present study aimed to evaluate the biological effects of the incorporation of highly pro-angiogenic copper (Cu) in 0106-B1-BG in vitro using human bone marrow-derived mesenchymal stromal cells (BMSCs) as well as its in vivo potential for bone regeneration. CuO was added to 0106-B1-BG in exchange for CaO, resulting in Cu-doped BG compositions containing 1.0, 2.5 and 5.0 wt % CuO (composition in wt %: 37.5 SiO2, 21.6/ 20.1/17.6 CaO, 5.9 Na2O, 4.0 P2O5, 12.0 K2O, 5.5 MgO, 12.5 B2O3, and 1.0/ 2.5/ 5.0 CuO). In vitro, the BGs' impact on the viability, proliferation, and growth patterns of BMSCs was evaluated. Analyses of protein secretion, matrix formation, and gene expression were used for the assessment of the BGs' influence on BMSCs regarding osteogenic differentiation and angiogenic stimulation. The presence of Cu improved cytocompatibility, osteogenic differentiation, and angiogenic response when compared with unmodified 0106-B1-BG in vitro. In vivo, a critical-size femoral defect in rats was filled with scaffolds made from BGs. Bone regeneration was evaluated by micro-computed tomography. Histological analysis was performed to assess bone maturation and angiogenesis. In vivo effects regarding defect closure, presence of osteoclastic cells or vascular structures in the defect were not significantly changed by the addition of Cu compared with undoped 0106-B1-BG scaffolds. Hence, while the in vitro properties of the 0106-B1-BG were significantly improved by the incorporation of Cu, further evaluation of the BG composition is necessary to transfer these effects to an in vivo setting.
Collapse
Affiliation(s)
- Linn Anna Fiehn
- Department of Orthopaedics, Heidelberg University Hospital, Heidelberg, Germany
| | - Elke Kunisch
- Department of Orthopaedics, Heidelberg University Hospital, Heidelberg, Germany
| | - Merve Saur
- Department of Orthopaedics, Heidelberg University Hospital, Heidelberg, Germany
| | | | - Christian Merle
- Department of Orthopaedics, Heidelberg University Hospital, Heidelberg, Germany
| | - Sébastien Hagmann
- Department of Orthopaedics, Heidelberg University Hospital, Heidelberg, Germany
| | - Adrian Stiller
- Johan Gadolin Process Chemistry Centre, Åbo Akademi University, Turku, Finland
| | - Leena Hupa
- Johan Gadolin Process Chemistry Centre, Åbo Akademi University, Turku, Finland
| | - Hana Kaňková
- Centre for Functional and Surface Functionalized Glass, Alexander Dubček University of Trenčín, Trenčín, Slovakia
| | - Dagmar Galusková
- Centre for Functional and Surface Functionalized Glass, Alexander Dubček University of Trenčín, Trenčín, Slovakia
| | - Tobias Renkawitz
- Department of Orthopaedics, Heidelberg University Hospital, Heidelberg, Germany
| | - Aldo R Boccaccini
- Institute of Biomaterials, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Fabian Westhauser
- Department of Orthopaedics, Heidelberg University Hospital, Heidelberg, Germany
| |
Collapse
|
2
|
Wang Y, Liu M, Zhang W, Liu H, Jin F, Mao S, Han C, Wang X. Mechanical strategies to promote vascularization for tissue engineering and regenerative medicine. BURNS & TRAUMA 2024; 12:tkae039. [PMID: 39350780 PMCID: PMC11441985 DOI: 10.1093/burnst/tkae039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 05/30/2024] [Accepted: 06/11/2024] [Indexed: 10/04/2024]
Abstract
Vascularization is a major challenge in the field of tissue engineering and regenerative medicine. Mechanical factors have been demonstrated to play a fundamental role in vasculogenesis and angiogenesis and can affect the architecture of the generated vascular network. Through the regulation of mechanical factors in engineered tissues, various mechanical strategies can be used to optimize the preformed vascular network and promote its rapid integration with host vessels. Optimization of the mechanical properties of scaffolds, including controlling scaffold stiffness, increasing surface roughness and anisotropic structure, and designing interconnected, hierarchical pore structures, is beneficial for the in vitro formation of vascular networks and the ingrowth of host blood vessels. The incorporation of hollow channels into scaffolds promotes the formation of patterned vascular networks. Dynamic stretching and perfusion can facilitate the formation and maturation of preformed vascular networks in vitro. Several indirect mechanical strategies provide sustained mechanical stimulation to engineered tissues in vivo, which further promotes the vascularization of implants within the body. Additionally, stiffness gradients, anisotropic substrates and hollow channels in scaffolds, as well as external cyclic stretch, boundary constraints and dynamic flow culture, can effectively regulate the alignment of vascular networks, thereby promoting better integration of prevascularized engineered tissues with host blood vessels. This review summarizes the influence and contribution of both scaffold-based and external stimulus-based mechanical strategies for vascularization in tissue engineering and elucidates the underlying mechanisms involved.
Collapse
Affiliation(s)
- Yiran Wang
- Department of Burns and Wound Care Center, The Second Affiliated Hospital of Zhejiang University College of Medicine, 88 Jiefang Road, Shangcheng District, Hangzhou 310009, China
- The Key Laboratory of the Diagnosis and Treatment of Severe Trauma and Burn of Zhejiang Province, 88 Jiefang Road, Shangcheng District, Hangzhou 310009, China
| | - Meixuan Liu
- Department of Burns and Wound Care Center, The Second Affiliated Hospital of Zhejiang University College of Medicine, 88 Jiefang Road, Shangcheng District, Hangzhou 310009, China
- The Key Laboratory of the Diagnosis and Treatment of Severe Trauma and Burn of Zhejiang Province, 88 Jiefang Road, Shangcheng District, Hangzhou 310009, China
| | - Wei Zhang
- Department of Burns and Wound Care Center, The Second Affiliated Hospital of Zhejiang University College of Medicine, 88 Jiefang Road, Shangcheng District, Hangzhou 310009, China
- The Key Laboratory of the Diagnosis and Treatment of Severe Trauma and Burn of Zhejiang Province, 88 Jiefang Road, Shangcheng District, Hangzhou 310009, China
| | - Huan Liu
- Department of Burns and Wound Care Center, The Second Affiliated Hospital of Zhejiang University College of Medicine, 88 Jiefang Road, Shangcheng District, Hangzhou 310009, China
- The Key Laboratory of the Diagnosis and Treatment of Severe Trauma and Burn of Zhejiang Province, 88 Jiefang Road, Shangcheng District, Hangzhou 310009, China
| | - Fang Jin
- Department of Burns and Wound Care Center, The Second Affiliated Hospital of Zhejiang University College of Medicine, 88 Jiefang Road, Shangcheng District, Hangzhou 310009, China
- The Key Laboratory of the Diagnosis and Treatment of Severe Trauma and Burn of Zhejiang Province, 88 Jiefang Road, Shangcheng District, Hangzhou 310009, China
| | - Shulei Mao
- Department of Burns and Plastic Surgery, Quhua Hospital of Zhejiang, 62 Wenchang Road, Quhua, Quzhou 324004, China
| | - Chunmao Han
- Department of Burns and Wound Care Center, The Second Affiliated Hospital of Zhejiang University College of Medicine, 88 Jiefang Road, Shangcheng District, Hangzhou 310009, China
- The Key Laboratory of the Diagnosis and Treatment of Severe Trauma and Burn of Zhejiang Province, 88 Jiefang Road, Shangcheng District, Hangzhou 310009, China
| | - Xingang Wang
- Department of Burns and Wound Care Center, The Second Affiliated Hospital of Zhejiang University College of Medicine, 88 Jiefang Road, Shangcheng District, Hangzhou 310009, China
- The Key Laboratory of the Diagnosis and Treatment of Severe Trauma and Burn of Zhejiang Province, 88 Jiefang Road, Shangcheng District, Hangzhou 310009, China
| |
Collapse
|
3
|
Xu Z, Wang B, Huang R, Guo M, Han D, Yin L, Zhang X, Huang Y, Li X. Efforts to promote osteogenesis-angiogenesis coupling for bone tissue engineering. Biomater Sci 2024; 12:2801-2830. [PMID: 38683241 DOI: 10.1039/d3bm02017g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/01/2024]
Abstract
Repair of bone defects exceeding a critical size has been always a big challenge in clinical practice. Tissue engineering has exhibited great potential to effectively repair the defects with less adverse effect than traditional bone grafts, during which how to induce vascularized bone formation has been recognized as a critical issue. Therefore, recently many studies have been launched to attempt to promote osteogenesis-angiogenesis coupling. This review summarized comprehensively and explored in depth current efforts to ameliorate the coupling of osteogenesis and angiogenesis from four aspects, namely the optimization of scaffold components, modification of scaffold structures, loading strategies for bioactive substances, and employment tricks for appropriate cells. Especially, the advantages and the possible reasons for every strategy, as well as the challenges, were elaborated. Furthermore, some promising research directions were proposed based on an in-depth analysis of the current research. This paper will hopefully spark new ideas and approaches for more efficiently boosting new vascularized bone formations.
Collapse
Affiliation(s)
- Zhiwei Xu
- College of Lab Medicine, Hebei North University, Zhangjiakou 075000, China
| | - Bingbing Wang
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, 37 Xueyuan Rd, Haidian District, Beijing, 100083, China.
| | - Ruoyu Huang
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, 37 Xueyuan Rd, Haidian District, Beijing, 100083, China.
| | - Mengyao Guo
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, 37 Xueyuan Rd, Haidian District, Beijing, 100083, China.
| | - Di Han
- College of Lab Medicine, Hebei North University, Zhangjiakou 075000, China
| | - Lan Yin
- Key Laboratory of Advanced Materials of Ministry of Education, Tsinghua University, Beijing 100084, China
| | - Xiaoyun Zhang
- College of Lab Medicine, Hebei North University, Zhangjiakou 075000, China
| | - Yong Huang
- College of Lab Medicine, Hebei North University, Zhangjiakou 075000, China
| | - Xiaoming Li
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, 37 Xueyuan Rd, Haidian District, Beijing, 100083, China.
| |
Collapse
|
4
|
Bi M, Yang K, Yu T, Wu G, Li Q. Cell-based mechanisms and strategies of co-culture system both in vivo and vitro for bone tissue engineering. Biomed Pharmacother 2023; 169:115907. [PMID: 37984308 DOI: 10.1016/j.biopha.2023.115907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Revised: 11/15/2023] [Accepted: 11/16/2023] [Indexed: 11/22/2023] Open
Abstract
The lack of a functional vascular supply has been identified as a major challenge limiting the clinical introduction of stem cell-based bone tissue engineering (BTE) for the repair of large-volume bone defects (LVBD). Various approaches have been explored to improve the vascular supply in tissue-engineered constructs, and the development of strategies that could effectively induce the establishment of a functional vascular supply has become a major goal of BTE research. One of the state-of-the-art methods is to incorporate both angiogenic and osteogenic cells in co-culture systems. This review clarifies the key concepts involved, summarises the cell types and models used to date, and systematically evaluates their performance. We also discuss the cell-to-cell communication between these two cell types and the strategies explored in BTE constructs with angiogenic and osteogenic cells to optimise their functions. In addition, we outline unresolved issues and remaining obstacles that need to be overcome for further development in this field and eventual successful repair of LVBD.
Collapse
Affiliation(s)
- Mengning Bi
- Department of Prosthetic Dentistry, Stomatological Hospital and Dental School of Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, China; Department of Orthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral diseases; Shanghai Key Laboratory of Stomatology; Shanghai Research Institute of Stomatology Shanghai, China
| | - Kaiwen Yang
- Department of Prosthetic Dentistry, Stomatological Hospital and Dental School of Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, China; Department of Oral Surgery, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; Shanghai Key Laboratory of Stomatology &Shanghai Research Institute of Stomatology; National Clinical Research Center of Stomatology, Shanghai, China
| | - Tao Yu
- Department of Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Gang Wu
- Department of Oral and Maxillofacial Surgery/Pathology, Amsterdam UMC and Academic Center for Dentistry Amsterdam (ACTA), Vrije Universiteit Amsterdam (VU), Amsterdam Movement Science (AMS), Amsterdam, the Netherlands; Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam (UvA) and Vrije Universiteit Amsterdam (VU), Amsterdam, the Netherlands.
| | - Qiong Li
- Department of Prosthetic Dentistry, Stomatological Hospital and Dental School of Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, China.
| |
Collapse
|
5
|
Bone Sialoprotein Immobilized in Collagen Type I Enhances Angiogenesis In Vitro and In Ovo. Polymers (Basel) 2023; 15:polym15041007. [PMID: 36850289 PMCID: PMC9968013 DOI: 10.3390/polym15041007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 01/24/2023] [Accepted: 02/14/2023] [Indexed: 02/22/2023] Open
Abstract
Bone fracture healing is a multistep process, including early immunological reactions, osteogenesis, and as a key factor, angiogenesis. Molecules inducing osteogenesis as well as angiogenesis are rare, but hold promise to be employed in bone tissue engineering. It has been demonstrated that the bone sialoprotein (BSP) can induce bone formation when immobilized in collagen type I, but its effect on angiogenesis still has to be characterized in detail. Therefore, the aim of this study was to analyse the effects of BSP immobilized in a collagen type I gel on angiogenesis. First, in vitro analyses with endothelial cells (HUVECs) were performed detecting enhancing effects of BSP on proliferation and gene expression of endothelial markers. A spheroid model was employed confirming these results. Finally, the inducing impact of BSP-collagen on vascular density was proved in a yolk sac membrane assay. Our results demonstrate that BSP is capable of inducing angiogenesis and confirm that collagen type I is the optimal carrier for this protein. Taking into account former results, and literature showing that BSP also induces osteogenesis, one can hypothesize that BSP couples angiogenesis and osteogenesis, making it a promising molecule to be used in bone tissue regeneration.
Collapse
|
6
|
Liu W, Feng Y, Wang X, Ding J, Li H, Guan H, Chen Z. Human umbilical vein endothelial cells-derived exosomes enhance cardiac function after acute myocardial infarction by activating the PI3K/AKT signaling pathway. Bioengineered 2022; 13:8850-8865. [PMID: 35361041 PMCID: PMC9161948 DOI: 10.1080/21655979.2022.2056317] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Currently, acute myocardial infarction (AMI) is one of the leading causes of human health issues worldwide. The sudden and continuous occlusion of the coronary artery results in myocardial hypoxic-ischemic necrosis, which is accompanied by inflammatory infiltration and fibrosis, leading to pathological cardiac remodeling. Exosome-based therapy is a promising cell-free approach for repairing the ischemic myocardium. This study aimed to explore the effects and mechanism of human umbilical vein endothelial cells (HUVECs)-derived exosomes on AMI. The results indicated that the localized injection of HUVECs-derived exosomes in the infarcted area could significantly improve cardiac function in AMI mouse models. It could also ameliorate myocardial fibrosis and decrease infarct size after AMI. Additionally, HUVECs-derived exosomes had cardioprotective effects on the H9C2 cells in hypoxic culture conditions, including increased cell viability and decreased lactate dehydrogenase (LDH) release. In both the in-vivo and in-vitro experiments, HUVECs-derived exosomes could effectively inhibit cardiomyocyte apoptosis. The low expression levels of Bcl-2–associated X protein (Bax) and cleaved caspase-3, high expression levels of B-cell lymphoma 2 (Bcl-2), phosphorylated phosphatidylinositol 3-kinase (p-PI3K), and phosphorylated protein kinase B (p-AKT) were detected in AMI mouse models treated with HUVECs-derived exosomes in-vivo. In conclusion, HUVECs-derived exosomes effectively enhanced cardiac function after AMI and inhibited cardiomyocyte apoptosis, which might be regulated through the phosphatidylinositol 3-kinase (PI3K)/ protein kinase B (AKT) signaling pathway.
Collapse
Affiliation(s)
- Wei Liu
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yu Feng
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xuehua Wang
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jiaxing Ding
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Huili Li
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Hongquan Guan
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Zhijian Chen
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
7
|
Liao Y, Fang Y, Zhu H, Huang Y, Zou G, Dai B, Rausch MA, Shi B. Concentrated Growth Factors Promote hBMSCs Osteogenic Differentiation in a Co-Culture System With HUVECs. Front Bioeng Biotechnol 2022; 10:837295. [PMID: 35387306 PMCID: PMC8979293 DOI: 10.3389/fbioe.2022.837295] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 02/09/2022] [Indexed: 01/01/2023] Open
Abstract
Osteogenesis is a complex physiologic process that occurs during bone regeneration. This process requires several growth factors that act on bone marrow-derived mesenchymal stem cells (BMSCs). Concentrated growth factor (CGF) is a new-generation platelet-rich derivative that is an appealing autologous material for application in tissue repair and bone regenerative medicine because it contains a variety of fibrin and growth factors. In this study, the effects of CGF on the proliferation and osteogenic differentiation of hBMSCs and human umbilical vein endothelial cells (HUVECs) were explored with in vitro cell co-culture experiments. HBMSCs and HUVECs were directly co-cultured at the ratio of 1:2 under different concentrations (0, 2, 5, 10, 20%) of CGF for 7 days. Alkaline phosphatase (ALP) staining and quantitative reverse transcription polymerase chain reaction were used to detect the effects of CGF on the expression of osteogenic genes (ALP, osteocalcin [OCN], type I collagen [COL-1], Runt-related transcription factor 2 [RUNX2]) and connexin 43 (CX43). RNA sequencing was used to explore potential regulated genes and signaling pathways that affect the osteogenesis of co-cultured hBMSCs exposed to CGF. The results showed higher expressions of ALP, COL-1, RUNX2, OCN, and CX43 in the direct co-culture group containing 10% CGF compared to the direct co-culture group without CGF and the indirect co-culture group. In summary, 10% CGF can significantly promote osteogenesis in hBMSCs directly co-cultured with HUVECs. Intercellular communication between the direct co-culture of hBMSCs and HUVECs through CX43 may be one of the main regulatory mechanisms.
Collapse
Affiliation(s)
- Yunyang Liao
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Laboratory of Facial Plastic and Reconstruction, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Oral Diseases, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Youran Fang
- Fujian Key Laboratory of Oral Diseases, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Hanghang Zhu
- Fujian Key Laboratory of Oral Diseases, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Yue Huang
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Laboratory of Facial Plastic and Reconstruction, Fujian Medical University, Fuzhou, China
| | - Gengsen Zou
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Laboratory of Facial Plastic and Reconstruction, Fujian Medical University, Fuzhou, China
| | - Bowen Dai
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Laboratory of Facial Plastic and Reconstruction, Fujian Medical University, Fuzhou, China
| | - Macro Aoqi Rausch
- Division of Orthodontics, University Clinic of Dentistry, Medical University of Vienna, Vienna, Austria
- *Correspondence: Macro Aoqi Rausch, ; Bin Shi,
| | - Bin Shi
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Laboratory of Facial Plastic and Reconstruction, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Oral Diseases, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
- *Correspondence: Macro Aoqi Rausch, ; Bin Shi,
| |
Collapse
|
8
|
Alvarez Echazú MI, Perna O, Olivetti CE, Antezana PE, Municoy S, Tuttolomondo MV, Galdopórpora JM, Alvarez GS, Olmedo DG, Desimone MF. Recent Advances in Synthetic and Natural Biomaterials-Based Therapy for Bone Defects. Macromol Biosci 2022; 22:e2100383. [PMID: 34984818 DOI: 10.1002/mabi.202100383] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 12/04/2021] [Indexed: 12/31/2022]
Abstract
Synthetic and natural biomaterials are a promising alternative for the treatment of critical-sized bone defects. Several parameters such as their porosity, surface, and mechanical properties are extensively pointed out as key points to recapitulate the bone microenvironment. Many biomaterials with this pursuit are employed to provide a matrix, which can supply the specific environment and architecture for an adequate bone growth. Nevertheless, some queries remain unanswered. This review discusses the recent advances achieved by some synthetic and natural biomaterials to mimic the native structure of bone and the manufacturing technology applied to obtain biomaterial candidates. The focus of this review is placed in the recent advances in the development of biomaterial-based therapy for bone defects in different types of bone. In this context, this review gives an overview of the potentialities of synthetic and natural biomaterials: polyurethanes, polyesters, hyaluronic acid, collagen, titanium, and silica as successful candidates for the treatment of bone defects.
Collapse
Affiliation(s)
- María I Alvarez Echazú
- Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET). Instituto de Química y Metabolismo del Fármaco (IQUIMEFA), Facultad de Farmacia y Bioquímica, Junín 956, Piso 3°, (1113) Buenos Aires, Argentina., Universidad de Buenos Aires, Junín 956, Piso 3°, Buenos Aires, 1113, Argentina.,Universidad de Buenos Aires, Facultad de Odontología, Cátedra de Anatomía Patológica, Marcelo T. de Alvear 2142 (1122), CABA, Argentina
| | - Oriana Perna
- Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET). Instituto de Química y Metabolismo del Fármaco (IQUIMEFA), Facultad de Farmacia y Bioquímica, Junín 956, Piso 3°, (1113) Buenos Aires, Argentina., Universidad de Buenos Aires, Junín 956, Piso 3°, Buenos Aires, 1113, Argentina
| | - Christian E Olivetti
- Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET). Instituto de Química y Metabolismo del Fármaco (IQUIMEFA), Facultad de Farmacia y Bioquímica, Junín 956, Piso 3°, (1113) Buenos Aires, Argentina., Universidad de Buenos Aires, Junín 956, Piso 3°, Buenos Aires, 1113, Argentina
| | - Pablo E Antezana
- Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET). Instituto de Química y Metabolismo del Fármaco (IQUIMEFA), Facultad de Farmacia y Bioquímica, Junín 956, Piso 3°, (1113) Buenos Aires, Argentina., Universidad de Buenos Aires, Junín 956, Piso 3°, Buenos Aires, 1113, Argentina
| | - Sofia Municoy
- Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET). Instituto de Química y Metabolismo del Fármaco (IQUIMEFA), Facultad de Farmacia y Bioquímica, Junín 956, Piso 3°, (1113) Buenos Aires, Argentina., Universidad de Buenos Aires, Junín 956, Piso 3°, Buenos Aires, 1113, Argentina
| | - María V Tuttolomondo
- Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET). Instituto de Química y Metabolismo del Fármaco (IQUIMEFA), Facultad de Farmacia y Bioquímica, Junín 956, Piso 3°, (1113) Buenos Aires, Argentina., Universidad de Buenos Aires, Junín 956, Piso 3°, Buenos Aires, 1113, Argentina
| | - Juan M Galdopórpora
- Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET). Instituto de Química y Metabolismo del Fármaco (IQUIMEFA), Facultad de Farmacia y Bioquímica, Junín 956, Piso 3°, (1113) Buenos Aires, Argentina., Universidad de Buenos Aires, Junín 956, Piso 3°, Buenos Aires, 1113, Argentina
| | - Gisela S Alvarez
- Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET). Instituto de Química y Metabolismo del Fármaco (IQUIMEFA), Facultad de Farmacia y Bioquímica, Junín 956, Piso 3°, (1113) Buenos Aires, Argentina., Universidad de Buenos Aires, Junín 956, Piso 3°, Buenos Aires, 1113, Argentina
| | - Daniel G Olmedo
- Universidad de Buenos Aires, Facultad de Odontología, Cátedra de Anatomía Patológica, Marcelo T. de Alvear 2142 (1122), CABA, Argentina.,CONICET, Consejo Nacional de Investigaciones Científicas y Técnicas, Godoy Cruz 2290, Buenos Aires, 1425, Argentina
| | - Martín F Desimone
- Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET). Instituto de Química y Metabolismo del Fármaco (IQUIMEFA), Facultad de Farmacia y Bioquímica, Junín 956, Piso 3°, (1113) Buenos Aires, Argentina., Universidad de Buenos Aires, Junín 956, Piso 3°, Buenos Aires, 1113, Argentina
| |
Collapse
|
9
|
Kang F, Yi Q, Gu P, Dong Y, Zhang Z, Zhang L, Bai Y. Controlled growth factor delivery system with osteogenic-angiogenic coupling effect for bone regeneration. J Orthop Translat 2022; 31:110-125. [PMID: 34976731 PMCID: PMC8671819 DOI: 10.1016/j.jot.2021.11.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 11/01/2021] [Accepted: 11/15/2021] [Indexed: 11/19/2022] Open
Abstract
Objective Bone regeneration involves a coordinated cascade of events that are regulated by several cytokines and growth factors, among which bone morphogenic protein-2 (BMP-2), vascular endothelial growth factor (VEGF) and fibroblast growth factor-2 (FGF-2) play important roles. In this study, we investigated the effects of dual release of the three growth factors on bone regeneration in femur defects. Methods A composite consisting of Gelatin microparticles loaded with VEGF/FGF-2 and poly(lactic-co-glycolic acid)-poly(ethylene glycol)-carboxyl (PLGA-PEG-COOH) microparticles loaded with BMP-2 encapsulated in a nano hydroxyapatite-poly actic-co-glycolic acid (nHA-PLGA) scaffold was prepared for the dual release of the growth factors. Results On the 14th day, decreased release rate of BMP-2 compared with FGF-2 and VEGF was observed. However, after 14 days, compared to FGF-2 and VEGF, BMP-2 showed an increased release rate. Controlled dual release of BMP-2 and VEGF, FGF-2 resulted in a significant osteogenic differentiation of bone mesenchymal stem cells (BMSCs). Moreover, effects of the composite scaffold on functional connection of osteoblast-vascular cells during bone development were evaluated. The synergistic effects of dual delivery of growth factors were shown to promote the expression of VEGF in BMSCs. Increased secretion of VEGF from BMSCs promoted the proliferation and angiogenic differentiation of human umbilical vein endothelial cells (HUVECs) in the co-culture system. At 12 weeks after implantation, blood vessel and bone formation were analyzed by micro-CT and histology. The composite scaffold significantly promoted the formation of blood vessels and new bone in femur defects. Conclusions These findings demonstrate that dual delivery of angiogenic factors and osteogenic factors from Gelatin and PLGA-PEG-COOH microparticles-based composite scaffolds exerted an osteogenic-angiogenic coupling effect on bone regeneration. This approach will inform on the development of appropriate designs of high-performance bioscaffolds for bone tissue engineering.
Collapse
Affiliation(s)
- Fei Kang
- Department of Biomedical Materials Science, Third Military Medical University, Chongqing, 400038, China
- School of Pharmacy, Chongqing Medical University, Chongqing, 400016, China
| | - Qiying Yi
- School of Pharmacy, Chongqing Medical University, Chongqing, 400016, China
| | - Pengcheng Gu
- School of Pharmacy, Chongqing Medical University, Chongqing, 400016, China
| | - Yuhan Dong
- School of Pharmacy, Chongqing Medical University, Chongqing, 400016, China
| | - Ziyang Zhang
- School of Pharmacy, Chongqing Medical University, Chongqing, 400016, China
| | - Lijuan Zhang
- School of Pharmacy, Chongqing Medical University, Chongqing, 400016, China
| | - Yan Bai
- School of Pharmacy, Chongqing Medical University, Chongqing, 400016, China
- Corresponding author. School of Pharmacy, Chongqing Medical University, District of Yuzhong, Chongqing, 400016, PR China.
| |
Collapse
|
10
|
Shafiee S, Shariatzadeh S, Zafari A, Majd A, Niknejad H. Recent Advances on Cell-Based Co-Culture Strategies for Prevascularization in Tissue Engineering. Front Bioeng Biotechnol 2021; 9:745314. [PMID: 34900955 PMCID: PMC8655789 DOI: 10.3389/fbioe.2021.745314] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 11/02/2021] [Indexed: 12/14/2022] Open
Abstract
Currently, the fabrication of a functional vascular network to maintain the viability of engineered tissues is a major bottleneck in the way of developing a more advanced engineered construct. Inspired by vasculogenesis during the embryonic period, the in vitro prevascularization strategies have focused on optimizing communications and interactions of cells, biomaterial and culture conditions to develop a capillary-like network to tackle the aforementioned issue. Many of these studies employ a combination of endothelial lineage cells and supporting cells such as mesenchymal stem cells, fibroblasts, and perivascular cells to create a lumenized endothelial network. These supporting cells are necessary for the stabilization of the newly developed endothelial network. Moreover, to optimize endothelial network development without impairing biomechanical properties of scaffolds or differentiation of target tissue cells, several other factors, including target tissue, endothelial cell origins, the choice of supporting cell, culture condition, incorporated pro-angiogenic factors, and choice of biomaterial must be taken into account. The prevascularization method can also influence the endothelial lineage cell/supporting cell co-culture system to vascularize the bioengineered constructs. This review aims to investigate the recent advances on standard cells used in in vitro prevascularization methods, their co-culture systems, and conditions in which they form an organized and functional vascular network.
Collapse
Affiliation(s)
- Sepehr Shafiee
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Siavash Shariatzadeh
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ali Zafari
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Alireza Majd
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hassan Niknejad
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
11
|
Braun J, Eckes S, Kilb MF, Fischer D, Eßbach C, Rommens PM, Drees P, Schmitz K, Nickel D, Ritz U. Mechanical characterization of rose bengal and green light crosslinked collagen scaffolds for regenerative medicine. Regen Biomater 2021; 8:rbab059. [PMID: 34858633 PMCID: PMC8633790 DOI: 10.1093/rb/rbab059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 09/30/2021] [Accepted: 10/22/2021] [Indexed: 11/13/2022] Open
Abstract
Collagen is one of the most important biomaterials for tissue engineering approaches. Despite its excellent biocompatibility, it shows the non-negligible disadvantage of poor mechanical stability. Photochemical crosslinking with rose bengal and green light (RGX) is an appropriate method to improve this property. The development of collagen laminates is helpful for further adjustment of the mechanical properties as well as the controlled release of incorporated substances. In this study, we investigate the impact of crosslinking and layering of two different collagen scaffolds on the swelling behavior and mechanical behavior in micro tensile tests to obtain information on its wearing comfort (stiffness, strength and ductility). The mechanical stability of the collagen material after degradation due to cell contact is examined using thickness measurements. There is no linear increase or decrease due to layering homologous laminates. Unexpectedly, a decrease in elongation at break, Young's modulus and ultimate tensile strength are measured when the untreated monolayer is compared to the crosslinked one. Furthermore, we can detect a connection between stability and cell proliferation. The results show that with variation in number and type of layers, collagen scaffolds with tailored mechanical properties can be produced. Such a multi-layered structure enables the release of biomolecules into inner or outer layers for biomedical applications.
Collapse
Affiliation(s)
- Joy Braun
- Department of Orthopedics and Traumatology, BiomaTiCS, University Medical Center, Johannes Gutenberg University, Langenbeckstraße 1, Mainz 55131, Germany
| | - Stefanie Eckes
- Clemens-Schöpf-Institute of Organic Chemistry and Biochemistry, Technical University of Darmstadt, Alarich-Weiss-Straße 4, Darmstadt 64287, Germany
| | - Michelle Fiona Kilb
- Clemens-Schöpf-Institute of Organic Chemistry and Biochemistry, Technical University of Darmstadt, Alarich-Weiss-Straße 4, Darmstadt 64287, Germany
| | - Dirk Fischer
- Berufsakademie Sachsen-Staatliche Studienakademie Glauchau, University of Cooperative Education, Kopernikusstraße 51, Glauchau 08371, Germany
| | - Claudia Eßbach
- Berufsakademie Sachsen-Staatliche Studienakademie Glauchau, University of Cooperative Education, Kopernikusstraße 51, Glauchau 08371, Germany
| | - Pol Maria Rommens
- Department of Orthopedics and Traumatology, BiomaTiCS, University Medical Center, Johannes Gutenberg University, Langenbeckstraße 1, Mainz 55131, Germany
| | - Philipp Drees
- Department of Orthopedics and Traumatology, BiomaTiCS, University Medical Center, Johannes Gutenberg University, Langenbeckstraße 1, Mainz 55131, Germany
| | - Katja Schmitz
- Clemens-Schöpf-Institute of Organic Chemistry and Biochemistry, Technical University of Darmstadt, Alarich-Weiss-Straße 4, Darmstadt 64287, Germany
| | - Daniela Nickel
- Berufsakademie Sachsen-Staatliche Studienakademie Glauchau, University of Cooperative Education, Kopernikusstraße 51, Glauchau 08371, Germany
| | - Ulrike Ritz
- Department of Orthopedics and Traumatology, BiomaTiCS, University Medical Center, Johannes Gutenberg University, Langenbeckstraße 1, Mainz 55131, Germany
| |
Collapse
|
12
|
Jamalpoor Z, Taromi N. Pre-vascularization of biomimetic 3-D scaffolds via direct co-culture of human umbilical cord derived osteogenic and angiogenic progenitor cells. J Drug Deliv Sci Technol 2021. [DOI: 10.1016/j.jddst.2021.102703] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
13
|
Xu HZ, Su JS. Restoration of critical defects in the rabbit mandible using osteoblasts and vascular endothelial cells co-cultured with vascular stent-loaded nano-composite scaffolds. J Mech Behav Biomed Mater 2021; 124:104831. [PMID: 34555626 DOI: 10.1016/j.jmbbm.2021.104831] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 09/04/2021] [Accepted: 09/07/2021] [Indexed: 01/07/2023]
Abstract
The success of large bone defect repair with tissue engineering technology depends mainly on angiogenesis and osteogenesis. In this study, we prepared poly-caprolactone/nano-hydroxyapatite/beta-calcium phosphate (PCL/nHA/β-TCP) composite scaffolds loaded with poly-(lactic-co-glycolic acid)/nano-hydroxyapatite/collagen/heparin sodium (PLGA/nHA/Col/HS) nanofiber small vascular stent by electrospinning and hot press forming-particle leaching methods. Supramolecular electrostatic self-assembly technology was used to modify the surfaces of small vascular stents to aid in hydrophilicity and anticoagulation. The surfaces of composite scaffolds were modified with an Arg-Gly-Asp (RGD) short peptide by physical adsorption to supply cell adhesion sites. The scaffolds were then combined with rabbit bone marrow-derived osteoblasts (OBs) and rabbit bone marrow-derived vascular endothelial cells (RVECs) to construct large, biologically active vascularized tissue-engineered bone in vitro; this bone was then used to repair critical bone defects in rabbit mandibles. Mechanical and biocompatibility testing results showed that PCL/nHA/β-TCP composite scaffolds loaded with small vascular stents had good surface structure, mechanical properties, biocompatibility, and bone-regeneration induction potential. Twelve weeks after implantation, histological analysis and X-ray scans showed that the use of osteoblasts and vascular endothelial cells co-cultured with PCL/nHA/β-TCP scaffolds was sufficient to repair critical defects in rabbit mandibles.
Collapse
Affiliation(s)
- Hong Zhen Xu
- Department of Prosthodontics, Shanghai Stomatological Hospital, Fudan University, Shanghai, China
| | - Jian Sheng Su
- Department of Prosthodontics, School & Hospital of Stomatology, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Tongji University, Shanghai, China.
| |
Collapse
|
14
|
Whelan IT, Moeendarbary E, Hoey DA, Kelly DJ. Biofabrication of vasculature in microphysiological models of bone. Biofabrication 2021; 13. [PMID: 34034238 DOI: 10.1088/1758-5090/ac04f7] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 05/25/2021] [Indexed: 11/12/2022]
Abstract
Bone contains a dense network of blood vessels that are essential to its homoeostasis, endocrine function, mineral metabolism and regenerative functions. In addition, bone vasculature is implicated in a number of prominent skeletal diseases, and bone has high affinity for metastatic cancers. Despite vasculature being an integral part of bone physiology and pathophysiology, it is often ignored or oversimplified inin vitrobone models. However, 3D physiologically relevant vasculature can now be engineeredin vitro, with microphysiological systems (MPS) increasingly being used as platforms for engineering this physiologically relevant vasculature. In recent years, vascularised models of bone in MPSs systems have been reported in the literature, representing the beginning of a possible technological step change in how bone is modelledin vitro. Vascularised bone MPSs is a subfield of bone research in its nascency, however given the impact of MPSs has had inin vitroorgan modelling, and the crucial role of vasculature to bone physiology, these systems stand to have a substantial impact on bone research. However, engineering vasculature within the specific design restraints of the bone niche is significantly challenging given the different requirements for engineering bone and vasculature. With this in mind, this paper aims to serve as technical guidance for the biofabrication of vascularised bone tissue within MPS devices. We first discuss the key engineering and biological considerations for engineering more physiologically relevant vasculaturein vitrowithin the specific design constraints of the bone niche. We next explore emerging applications of vascularised bone MPSs, and conclude with a discussion on the current status of vascularised bone MPS biofabrication and suggest directions for development of next generation vascularised bone MPSs.
Collapse
|
15
|
Gurel Pekozer G, Abay Akar N, Cumbul A, Beyzadeoglu T, Torun Kose G. Investigation of Vasculogenesis Inducing Biphasic Scaffolds for Bone Tissue Engineering. ACS Biomater Sci Eng 2021; 7:1526-1538. [PMID: 33740374 DOI: 10.1021/acsbiomaterials.0c01071] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Vascularization is the main obstacle for the bone tissue engineering strategies since the defect size is generally large. Incorporation of angiogenic factors is one of the strategies employed in order to accelerate vascularization and improve bone healing. In this study, a biphasic scaffold consisting of fibrous poly(lactide-co-glycolide) (PLGA) and poly(lactide-co-glycolide)-block-poly(ethylene glycol)-block-poly(lactide-co-glycolide) (PLGA-PEG-PLGA) hydrogel loaded with vascular endothelial growth factor-A (VEGF) inducer, GS4012, was constructed. Mesenchymal stem cells isolated from rat bone marrow (rBMSCs) were used for differentiation into osteogenic cells, and endothelial cells isolated from rat peripheral blood (rPBECs) were used to test the in vitro endothelial cell recruitment. The biphasic scaffold was tested for cell proliferation, ALP expression, VEGF induction, expression of osteogenic genes by rBMSCs, and recruitment of rPBECs in vitro and for improved bone healing and vascularization in vivo on critical size rat cranial defects. Endothelial migration through porous insert and VEGF induction were obtained in vitro in response to GS4012 as well as the upregulation of ALP, Runx2, Col I, and OC gene expressions. The biphasic scaffold was also shown to be effective in improving endothelial cell recruitment, vascularization, and bone healing in vivo. Thus, the proposed design has a great potential for the healing of critical size bone defect in tissue engineering studies according to both in vitro and in vivo investigations.
Collapse
Affiliation(s)
- Gorke Gurel Pekozer
- Biomedical Engineering Department, Faculty of Electrical and Electronics Engineering, Yildiz Technical University, Istanbul 34220, Turkey
| | - Nergis Abay Akar
- Genetics and Bioengineering Department, Faculty of Engineering, Yeditepe University, Istanbul 34755, Turkey
| | - Alev Cumbul
- Histology and Embryology Department, Faculty of Medicine, Yeditepe University, Istanbul 34755, Turkey
| | - Tahsin Beyzadeoglu
- Orthopaedics and Traumatology, Facuty of Health Sciences, Halic University Beyzadeoglu Clinic, Istanbul 34738, Turkey
| | - Gamze Torun Kose
- Genetics and Bioengineering Department, Faculty of Engineering, Yeditepe University, Istanbul 34755, Turkey
| |
Collapse
|
16
|
Bellani C, Yue K, Flaig F, Hébraud A, Ray P, Annabi N, Selistre de Araújo HS, Branciforti MC, Minarelli Gaspar AM, Shin SR, Khademhosseini A, Schlatter G. Suturable elastomeric tubular grafts with patterned porosity for rapid vascularization of 3D constructs. Biofabrication 2021; 13. [PMID: 33482658 DOI: 10.1088/1758-5090/abdf1d] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 01/22/2021] [Indexed: 12/11/2022]
Abstract
Vascularization is considered to be one of the key challenges in engineering functional 3D tissues. Engineering suturable vascular grafts containing pores with diameter of several tens of microns in tissue engineered constructs may provide an instantaneous blood perfusion through the grafts improving cell infiltration and thus, allowing rapid vascularization and vascular branching. The aim of this work was to develop suturable tubular scaffolds to be integrated in biofabricated constructs, enabling the direct connection of the biofabricated construct with the host blood stream, providing an immediate blood flow inside the construct. Here, tubular grafts with customizable shapes (tubes, Y-shape capillaries) and controlled diameter ranging from several hundreds of microns to few mm are fabricated based on poly(glycerol sebacate) (PGS) / poly(vinyl alcohol) (PVA) electrospun scaffolds. Furthermore, a network of pore channels of diameter in the order of 100 µm was machined by laser femtosecond ablation in the tube wall. Both non-machined and laser machined tubular scaffolds elongated more than 100% of their original size have shown suture retention, being 5.85 and 3.96 N/mm2 respectively. To demonstrate the potential of application, the laser machined porous grafts were embedded in gelatin methacryloyl (GelMA) hydrogels, resulting in elastomeric porous tubular graft/GelMA 3D constructs. These constructs were then co-seeded with osteoblast-like cells (MG-63) at the external side of the graft and endothelial cells (HUVEC) inside, forming a bone osteon model. The laser machined pore network allowed an immediate endothelial cell flow towards the osteoblasts enabling the osteoblasts and endothelial cells to interact and form 3D structures. This rapid vascularization approach could be applied, not only for bone tissue regeneration, but also for a variety of tissues and organs.
Collapse
Affiliation(s)
- Caroline Bellani
- University of Sao Paulo, AVENIDA TRABALHADOR SÃO-CARLENSE, 400, Sao Carlos, São Paulo, 13566-590, BRAZIL
| | - Kan Yue
- South China Advanced Institute for Soft Matter Science and Technology, South China University of Technology, 381 Wushan Rd, Guangzhou, Guangdong, 510641, CHINA
| | - Florence Flaig
- ICPEES, University of Strasbourg, 25 rue Bécquerel, Strasbourg, 67087, FRANCE
| | - Anne Hébraud
- ICPEES, 25 rue Bécquerel, Strasbourg, 67087, FRANCE
| | - Pengfei Ray
- Division of Health Sciences and Technology, MIT, 45 Carleton Street, Cambridge, Massachusetts, 02142, UNITED STATES
| | - Nasim Annabi
- Department of Chemical and Biomolecular Engineering, UCLA, 5531 Boelter Hall, Los Angeles, California, CA 90095, UNITED STATES
| | | | - Marcia Cristina Branciforti
- Depatament of Materials Engineering, University of Sao Paulo, AVENIDA TRABALHADOR SÃO-CARLENSE, 400, ARNOLD SCHMITED, SAO CARLOS, Sao Paulo, SAO PAULO, 13566-590, BRAZIL
| | - Ana Maria Minarelli Gaspar
- Department of Morphology, School of Dentistry at Araraquara, Sao Paulo State University Julio de Mesquita Filho, R. Humaitá, 1680, Araraquara, SP, 14801-385, BRAZIL
| | - Su Ryon Shin
- Medicine, Harvard Medical School, 25 Shattuck Street, Boston, Massachusetts, MA 02115, UNITED STATES
| | - Ali Khademhosseini
- Department of Chemical and Biomolecular Engineering, UCLA, 5531 Boelter Hall, Los Angeles, California, CA 90095, UNITED STATES
| | - Guy Schlatter
- ICPEES, University of Strasbourg, 25 rue Bécquerel, Strasbourg, 67087, FRANCE
| |
Collapse
|
17
|
3D printing of tissue engineering scaffolds: a focus on vascular regeneration. Biodes Manuf 2021; 4:344-378. [PMID: 33425460 PMCID: PMC7779248 DOI: 10.1007/s42242-020-00109-0] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Accepted: 10/24/2020] [Indexed: 01/31/2023]
Abstract
Tissue engineering is an emerging means for resolving the problems of tissue repair and organ replacement in regenerative medicine. Insufficient supply of nutrients and oxygen to cells in large-scale tissues has led to the demand to prepare blood vessels. Scaffold-based tissue engineering approaches are effective methods to form new blood vessel tissues. The demand for blood vessels prompts systematic research on fabrication strategies of vascular scaffolds for tissue engineering. Recent advances in 3D printing have facilitated fabrication of vascular scaffolds, contributing to broad prospects for tissue vascularization. This review presents state of the art on modeling methods, print materials and preparation processes for fabrication of vascular scaffolds, and discusses the advantages and application fields of each method. Specially, significance and importance of scaffold-based tissue engineering for vascular regeneration are emphasized. Print materials and preparation processes are discussed in detail. And a focus is placed on preparation processes based on 3D printing technologies and traditional manufacturing technologies including casting, electrospinning, and Lego-like construction. And related studies are exemplified. Transformation of vascular scaffolds to clinical application is discussed. Also, four trends of 3D printing of tissue engineering vascular scaffolds are presented, including machine learning, near-infrared photopolymerization, 4D printing, and combination of self-assembly and 3D printing-based methods.
Collapse
|
18
|
Ehnert S, Rinderknecht H, Aspera-Werz RH, Häussling V, Nussler AK. Use of in vitro bone models to screen for altered bone metabolism, osteopathies, and fracture healing: challenges of complex models. Arch Toxicol 2020; 94:3937-3958. [PMID: 32910238 PMCID: PMC7655582 DOI: 10.1007/s00204-020-02906-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 09/03/2020] [Indexed: 02/06/2023]
Abstract
Approx. every third hospitalized patient in Europe suffers from musculoskeletal injuries or diseases. Up to 20% of these patients need costly surgical revisions after delayed or impaired fracture healing. Reasons for this are the severity of the trauma, individual factors, e.g, the patients' age, individual lifestyle, chronic diseases, medication, and, over 70 diseases that negatively affect the bone quality. To investigate the various disease constellations and/or develop new treatment strategies, many in vivo, ex vivo, and in vitro models can be applied. Analyzing these various models more closely, it is obvious that many of them have limits and/or restrictions. Undoubtedly, in vivo models most completely represent the biological situation. Besides possible species-specific differences, ethical concerns may question the use of in vivo models especially for large screening approaches. Challenging whether ex vivo or in vitro bone models can be used as an adequate replacement for such screenings, we here summarize the advantages and challenges of frequently used ex vivo and in vitro bone models to study disturbed bone metabolism and fracture healing. Using own examples, we discuss the common challenge of cell-specific normalization of data obtained from more complex in vitro models as one example of the analytical limits which lower the full potential of these complex model systems.
Collapse
Affiliation(s)
- Sabrina Ehnert
- Siegfried Weller Research Institute at the BG Trauma Center Tübingen, Department of Trauma and Reconstructive Surgery, University of Tübingen, Tübingen, Germany.
| | - Helen Rinderknecht
- Siegfried Weller Research Institute at the BG Trauma Center Tübingen, Department of Trauma and Reconstructive Surgery, University of Tübingen, Tübingen, Germany
| | - Romina H Aspera-Werz
- Siegfried Weller Research Institute at the BG Trauma Center Tübingen, Department of Trauma and Reconstructive Surgery, University of Tübingen, Tübingen, Germany
| | - Victor Häussling
- Siegfried Weller Research Institute at the BG Trauma Center Tübingen, Department of Trauma and Reconstructive Surgery, University of Tübingen, Tübingen, Germany
| | - Andreas K Nussler
- Siegfried Weller Research Institute at the BG Trauma Center Tübingen, Department of Trauma and Reconstructive Surgery, University of Tübingen, Tübingen, Germany
| |
Collapse
|
19
|
Eckes S, Braun J, Wack JS, Ritz U, Nickel D, Schmitz K. Rose Bengal Crosslinking to Stabilize Collagen Sheets and Generate Modulated Collagen Laminates. Int J Mol Sci 2020; 21:E7408. [PMID: 33049938 PMCID: PMC7582313 DOI: 10.3390/ijms21197408] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Revised: 10/05/2020] [Accepted: 10/07/2020] [Indexed: 01/07/2023] Open
Abstract
For medical application, easily accessible biomaterials with tailored properties are desirable. Collagen type I represents a biomaterial of choice for regenerative medicine and tissue engineering. Here, we present a simple method to modify the properties of collagen and to generate collagen laminates. We selected three commercially available collagen sheets with different thicknesses and densities and examined the effect of rose bengal and green light collagen crosslinking (RGX) on properties such as microstructure, swelling degree, mechanical stability, cell compatibility and drug release. The highest impact of RGX was measured for Atelocollagen, for which the swelling degree was reduced from 630% (w/w) to 520% (w/w) and thickness measured under force application increased from 0.014 mm to 0.455 mm, indicating a significant increase in mechanical stability. Microstructural analysis revealed that the sponge-like structure was replaced by a fibrous structure. While the initial burst effect during vancomycin release was not influenced by crosslinking, RGX increased cell proliferation on sheets of Atelocollagen and on Collagen Solutions. We furthermore demonstrate that RGX can be used to covalently attach different sheets to create materials with combined properties, making the modification and combination of readily available sheets with RGX an attractive approach for clinical application.
Collapse
Affiliation(s)
- Stefanie Eckes
- Clemens-Schöpf-Institute of Organic Chemistry and Biochemistry, Technical University of Darmstadt, Alarich-Weiss-Straße 8, 64287 Darmstadt, Germany
| | - Joy Braun
- Department of Orthopaedics and Traumatology, BiomaTiCS, University Medical Center, Johannes Gutenberg University, Langenbeckstraße 1, 55131 Mainz, Germany
| | - Julia S Wack
- Clemens-Schöpf-Institute of Organic Chemistry and Biochemistry, Technical University of Darmstadt, Alarich-Weiss-Straße 8, 64287 Darmstadt, Germany
| | - Ulrike Ritz
- Department of Orthopaedics and Traumatology, BiomaTiCS, University Medical Center, Johannes Gutenberg University, Langenbeckstraße 1, 55131 Mainz, Germany
| | - Daniela Nickel
- Berufsakademie Sachsen-Staatliche Studienakademie Glauchau, University of Cooperative Education, Kopernikusstraße 51, 08371 Glauchau, Germany
| | - Katja Schmitz
- Clemens-Schöpf-Institute of Organic Chemistry and Biochemistry, Technical University of Darmstadt, Alarich-Weiss-Straße 8, 64287 Darmstadt, Germany
| |
Collapse
|
20
|
Cell seeding accelerates the vascularization of tissue engineering constructs in hypertensive mice. Hypertens Res 2020; 44:23-35. [PMID: 32778779 DOI: 10.1038/s41440-020-0524-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 06/22/2020] [Accepted: 06/24/2020] [Indexed: 11/08/2022]
Abstract
Rapid blood vessel ingrowth into transplanted constructs represents the key requirement for successful tissue engineering. Seeding three-dimensional scaffolds with suitable cells is an approved technique for this challenge. Since a plethora of patients suffer from widespread diseases that limit the capacity of neoangiogenesis (e.g., hypertension), we investigated the incorporation of cell-seeded poly-L-lactide-co-glycolide scaffolds in hypertensive (BPH/2J, group A) and nonhypertensive (BPN/3J, group B) mice. Collagen-coated scaffolds (A1 and B1) were additionally seeded with osteoblast-like (A2 and B2) and mesenchymal stem cells (A3 and B3). After implantation into dorsal skinfold chambers, inflammation and newly formed microvessels were measured using repetitive intravital fluorescence microscopy for 2 weeks. Apart from a weak inflammatory response in all groups, significantly increased microvascular densities were found in cell-seeded scaffolds (day 14, A2: 192 ± 12 cm/cm2, A3: 194 ± 10 cm/cm2, B2: 249 ± 19 cm/cm2, B3: 264 ± 17 cm/cm2) when compared with controls (A1: 129 ± 10 cm/cm2, B1: 185 ± 8 cm/cm2). In this context, hypertensive mice showed reduced neoangiogenesis in comparison with nonhypertensive animals. Therefore, seeding approved scaffolds with organ-specific or pluripotent cells is a very promising technique for tissue engineering in hypertensive organisms.
Collapse
|
21
|
Braun J, Eckes S, Rommens PM, Schmitz K, Nickel D, Ritz U. Toxic Effect of Vancomycin on Viability and Functionality of Different Cells Involved in Tissue Regeneration. Antibiotics (Basel) 2020; 9:E238. [PMID: 32397094 PMCID: PMC7277215 DOI: 10.3390/antibiotics9050238] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 05/01/2020] [Accepted: 05/03/2020] [Indexed: 01/10/2023] Open
Abstract
To prevent infections local delivery of antibiotics is a useful tool. Especially in bone fractures, vancomycin impregnated bone cements are often used allowing high concentrations of antibiotics at the infection side without high serum concentrations. However, besides potential pathogens, cells involved in tissue regeneration may also be affected by the drug. We investigated the effect of vancomycin on the viability and functionality on osteoblasts, endothelial cells, fibroblasts and skeletal muscle cells. Our results show that the viability of all cells analyzed was reduced by vancomycin and that the observed effects were time and concentration dependent. The most pronounced toxic effect was detected on day three when even the lowest concentration of 0.01 mg/ml led to a significant decrease in proliferation compared to control. Functionality assays of osteoblasts and skeletal muscle cells revealed a sensitive reaction of the cells to the drug, indicating that vancomycin is toxic to these cells during the process of differentiation. These data suggest that the vancomycin administration is critical for cell survival and function. Therefore, the concentration of administered antibiotics needs to be carefully evaluated to find a balance between defense against pathogens and functionality of host cells and tissues.
Collapse
Affiliation(s)
- Joy Braun
- Department of Orthopaedics and Traumatology, BiomaTiCS, University Medical Center, Johannes Gutenberg University, Langenbeckstraße 1, 55131 Mainz, Germany; (J.B.); (P.M.R.)
| | - Stefanie Eckes
- Clements-Schöpf-Institute of Organic Chemistry and Biochemistry, Technische Universität Darmstadt, Alarich-Weiss-Straße 4, 64287 Darmstadt, Germany; (S.E.); (K.S.)
| | - Pol Maria Rommens
- Department of Orthopaedics and Traumatology, BiomaTiCS, University Medical Center, Johannes Gutenberg University, Langenbeckstraße 1, 55131 Mainz, Germany; (J.B.); (P.M.R.)
| | - Katja Schmitz
- Clements-Schöpf-Institute of Organic Chemistry and Biochemistry, Technische Universität Darmstadt, Alarich-Weiss-Straße 4, 64287 Darmstadt, Germany; (S.E.); (K.S.)
| | - Daniela Nickel
- Berufsakademie-Sachsen—Staatliche Studienakademie Glauchau, University of Cooperative Education, Kopernikusstraße 51, 08371 Glauchau, Germany;
| | - Ulrike Ritz
- Department of Orthopaedics and Traumatology, BiomaTiCS, University Medical Center, Johannes Gutenberg University, Langenbeckstraße 1, 55131 Mainz, Germany; (J.B.); (P.M.R.)
| |
Collapse
|
22
|
Nagai M, Kato K, Soga S, Santra TS, Shibata T. Scalable Parallel Manipulation of Single Cells Using Micronozzle Array Integrated with Bidirectional Electrokinetic Pumps. MICROMACHINES 2020; 11:mi11040442. [PMID: 32331468 PMCID: PMC7231381 DOI: 10.3390/mi11040442] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Revised: 04/17/2020] [Accepted: 04/22/2020] [Indexed: 12/27/2022]
Abstract
High throughput reconstruction of in vivo cellular environments allows for efficient investigation of cellular functions. If one-side-open multi-channel microdevices are integrated with micropumps, the devices will achieve higher throughput in the manipulation of single cells while maintaining flexibility and open accessibility. This paper reports on the integration of a polydimethylsiloxane (PDMS) micronozzle array and bidirectional electrokinetic pumps driven by DC-biased AC voltages. Pt/Ti and indium tin oxide (ITO) electrodes were used to study the effect of DC bias and peak-to-peak voltage and electrodes in a low conductivity isotonic solution. The flow was bidirectionally controlled by changing the DC bias. A pump integrated with a micronozzle array was used to transport single HeLa cells into nozzle holes. The application of DC-biased AC voltage (100 kHz, 10 Vpp, and VDC: -4 V) provided a sufficient electroosmotic flow outside the nozzle array. This integration method of nozzle and pumps is anticipated to be a standard integration method. The operating conditions of DC-biased AC electrokinetic pumps in a biological buffer was clarified and found useful for cell manipulation.
Collapse
Affiliation(s)
- Moeto Nagai
- Department of Mechanical Engineering, Toyohashi University of Technology, Toyohashi, Aichi 441-8580, Japan; (K.K.); (S.S.); (T.S.)
- Correspondence: ; Tel.: +81-532-44-6701
| | - Keita Kato
- Department of Mechanical Engineering, Toyohashi University of Technology, Toyohashi, Aichi 441-8580, Japan; (K.K.); (S.S.); (T.S.)
| | - Satoshi Soga
- Department of Mechanical Engineering, Toyohashi University of Technology, Toyohashi, Aichi 441-8580, Japan; (K.K.); (S.S.); (T.S.)
| | - Tuhin Subhra Santra
- Department of Engineering Design, Indian Institute of Technology Madras, Tamil Nadu 600036, India;
| | - Takayuki Shibata
- Department of Mechanical Engineering, Toyohashi University of Technology, Toyohashi, Aichi 441-8580, Japan; (K.K.); (S.S.); (T.S.)
| |
Collapse
|
23
|
Shahabipour F, Oskuee RK, Dehghani H, Shokrgozar MA, Aninwene GE, Bonakdar S. Cell-cell interaction in a coculture system consisting of CRISPR/Cas9 mediated GFP knock-in HUVECs and MG-63 cells in alginate-GelMA based nanocomposites hydrogel as a 3D scaffold. J Biomed Mater Res A 2020; 108:1596-1606. [PMID: 32180319 DOI: 10.1002/jbm.a.36928] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2019] [Revised: 03/05/2020] [Accepted: 03/09/2020] [Indexed: 12/12/2022]
Abstract
The interaction between osteogenic and angiogenic cells through a coculturing system in biocompatible materials has been considered for successfully engineering vascularized bone tissue equivalents. In this study, we developed a hydrogel-blended scaffold consisted of gelatin methacryloyl (GelMA) and alginate enriched with hydroxyapatite nanoparticles (HAP) to model an in vitro prevascularized bone construct. The hydrogel-based scaffold revealed a higher mechanical stiffness than those of pure (GelMA), alginate, and (GelMA+ HAP) hydrogels. In the present study, we generated a green fluorescent protein (GFP) knock-in umbilical vein endothelial cells (HUVECs) cell line using the CRISPR/Cas9 technology. The GFP was inserted into the human-like ROSA locus of HUVECs genome. HUVECs expressing GFP were cocultured with OB-like cells (MG-63) within three-dimensionally (3D) fabricated hydrogel to investigate the response of cocultured osteoblasts and endothelial cells in a 3D structure. Cell viability under the 3D cocultured gel was higher than the 3D monocultured. Compared to the 3D monocultured condition, the cells were aligned and developed into the vessel-like structures. During 14 days of culture periods, the cells displayed actin protrusions by the formation of spike-like filopodia in the 3D cocultured model. Angiogenic and osteogenic-related genes such as CD31, vWF, and osteocalcin showed higher expression in the cocultured versus the monocultured. These results have collectively indicated that the 3D cocultured hydrogel facilitates interaction among cells, thereby having a greater effect on angiogenic and osteogenic properties in the absence of induction media.
Collapse
Affiliation(s)
| | - Reza K Oskuee
- Targeted Drug Delivery Research Center, Institute of Pharmaceutical Technology, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hesam Dehghani
- Division of Biotechnology, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, Mashhad, Iran.,Department of Basic Science, Faculty of Veterinary medicine, Ferdowsi University of Mashhad, Mashhad, Iran
| | | | - George E Aninwene
- Department of Bioengineering, University of California-Los Angeles, Los Angeles, California, USA.,Center for Minimally Invasive Therapeutics (C-MIT), University of California-Los Angeles, Los Angeles, California, USA
| | - Shahin Bonakdar
- National Cell Bank of Iran, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
24
|
Tian X, Yuan X, Feng D, Wu M, Yuan Y, Ma C, Xie D, Guo J, Liu C, Lu Z. In vivo study of polyurethane and tannin-modified hydroxyapatite composites for calvarial regeneration. J Tissue Eng 2020; 11:2041731420968030. [PMID: 33282174 PMCID: PMC7682243 DOI: 10.1177/2041731420968030] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 10/02/2020] [Indexed: 12/20/2022] Open
Abstract
Biomaterial mediated bone regeneration is an attractive strategy for bone defect treatment. Organic/inorganic composites have been well established as effective bone graft. Here, the bone regenerative effect of the composites made from tannic acid (TA) modified hydroxyapatite (HA) (THA) or TA & silver nanoparticles (Ag NPs) modified HA (Ag-THA) and polyurethane (PU) was evaluated on critical-sized calvarial defects in rats. The in vivo study indicates that PU/THA and PU/Ag-THA scaffolds exhibited acceptable biocompatibility and induced significantly enhanced bone mineral densities comparing with the blank control (CON) group as well as PU/HA group. The inclusion of TA on HA brought the composites with enhanced osteogenesis and angiogenesis, evidenced by osteocalcin (OCN) and vascular endothelial growth factor (VEGF) immunohistochemical staining. Tartrate resistant acid phosphatase (TRAP) staining showed high osteoclast activity along with osteogenesis, especially in PU/THA and PU/Ag-THA groups. However, further introduction of Ag NPs on HA depressed the angiogenesis of the composites, leading to even lower VEGF expression than that of CON group. This study once more proved that THA can serve as a better bone composite component that pure HA and can promote osteogenesis and angiogenesis. While, the introduction of antimicrobial Ag NPs on HA need to be controlled in some extent not to affect the angiogenesis of the composites.
Collapse
Affiliation(s)
- Xinggui Tian
- Department of Histology and Embryology, School of Basic Medical Sciences, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Southern Medical University, Guangzhou, China
- Department of Orthopaedics, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, P. R. China
| | - Xiaowei Yuan
- Department of Histology and Embryology, School of Basic Medical Sciences, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Southern Medical University, Guangzhou, China
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Daxiong Feng
- Department of Orthopaedics, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, P. R. China
| | - Min Wu
- Department of Histology and Embryology, School of Basic Medical Sciences, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Southern Medical University, Guangzhou, China
| | - Yuping Yuan
- Department of Material Science and Engineering, Southern University of Science and Technology, Shenzhen, China
| | | | - Denghui Xie
- Department of Histology and Embryology, School of Basic Medical Sciences, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Southern Medical University, Guangzhou, China
| | - Jinshan Guo
- Department of Histology and Embryology, School of Basic Medical Sciences, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Southern Medical University, Guangzhou, China
| | - Chao Liu
- Aleo BME, Inc., State College, PA, USA
| | - Zhihui Lu
- Department of Histology and Embryology, School of Basic Medical Sciences, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Southern Medical University, Guangzhou, China
| |
Collapse
|
25
|
Taniguchi D, Matsumoto K, Machino R, Takeoka Y, Elgalad A, Taura Y, Oyama S, Tetsuo T, Moriyama M, Takagi K, Kunizaki M, Tsuchiya T, Miyazaki T, Hatachi G, Matsuo N, Nakayama K, Nagayasu T. Human lung microvascular endothelial cells as potential alternatives to human umbilical vein endothelial cells in bio-3D-printed trachea-like structures. Tissue Cell 2019; 63:101321. [PMID: 32223949 DOI: 10.1016/j.tice.2019.101321] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 11/29/2019] [Accepted: 12/02/2019] [Indexed: 11/28/2022]
Abstract
BACKGROUND We have been trying to produce scaffold-free structures for airway regeneration using a bio-3D-printer with spheroids, to avoid scaffold-associated risks such as infection. Previous studies have shown that human umbilical vein endothelial cells (HUVECs) play an important role in such structures, but HUVECs cannot be isolated from adult humans. The aim of this study was to identify alternatives to HUVECs for use in scaffold-free structures. METHODS Three types of structure were compared, made of chondrocytes and mesenchymal stem cells with HUVECs, human lung microvascular endothelial cells (HMVEC-Ls), and induced pluripotent stem cell (iPSC)-derived endothelial cells. RESULTS No significant difference in tensile strength was observed between the three groups. Histologically, some small capillary-like tube formations comprising CD31-positive cells were observed in all groups. The number and diameters of such formations were significantly lower in the iPSC-derived endothelial cell group than in other groups. Glycosaminoglycan content was significantly lower in the iPSC-derived endothelial cell group than in the HUVEC group, while no significant difference was observed between the HUVEC and HMVEC-L groups. CONCLUSIONS HMVEC-Ls can replace HUVECs as a cell source for scaffold-free trachea-like structures. However, some limitations were associated with iPSC-derived endothelial cells.
Collapse
Affiliation(s)
- D Taniguchi
- Department of Surgical Oncology, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan; Medical-engineering Hybrid Professional Development Program, Nagasaki University, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
| | - K Matsumoto
- Department of Surgical Oncology, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan; Medical-engineering Hybrid Professional Development Program, Nagasaki University, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan.
| | - R Machino
- Department of Surgical Oncology, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
| | - Y Takeoka
- Department of Surgical Oncology, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan; Medical-engineering Hybrid Professional Development Program, Nagasaki University, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
| | - A Elgalad
- Department of Surgical Oncology, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan; Medical-engineering Hybrid Professional Development Program, Nagasaki University, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
| | - Y Taura
- Department of Surgical Oncology, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
| | - S Oyama
- Department of Surgical Oncology, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan; Medical-engineering Hybrid Professional Development Program, Nagasaki University, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
| | - T Tetsuo
- Department of Surgical Oncology, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan; Medical-engineering Hybrid Professional Development Program, Nagasaki University, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
| | - M Moriyama
- Department of Surgical Oncology, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan; Medical-engineering Hybrid Professional Development Program, Nagasaki University, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
| | - K Takagi
- Medical-engineering Hybrid Professional Development Program, Nagasaki University, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
| | - M Kunizaki
- Department of Surgical Oncology, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
| | - T Tsuchiya
- Department of Surgical Oncology, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
| | - T Miyazaki
- Department of Surgical Oncology, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
| | - G Hatachi
- Department of Surgical Oncology, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan; Medical-engineering Hybrid Professional Development Program, Nagasaki University, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
| | - N Matsuo
- Department of Surgical Oncology, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan; Medical-engineering Hybrid Professional Development Program, Nagasaki University, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
| | - K Nakayama
- Department of Regenerative Medicine and Biomedical Engineering Faculty of Medicine, Saga University, 1 Honjocho, Saga, 840-8502, Japan
| | - T Nagayasu
- Department of Surgical Oncology, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan; Medical-engineering Hybrid Professional Development Program, Nagasaki University, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
| |
Collapse
|
26
|
Ferreira FV, Souza LP, Martins TMM, Lopes JH, Mattos BD, Mariano M, Pinheiro IF, Valverde TM, Livi S, Camilli JA, Goes AM, Gouveia RF, Lona LMF, Rojas OJ. Nanocellulose/bioactive glass cryogels as scaffolds for bone regeneration. NANOSCALE 2019; 11:19842-19849. [PMID: 31441919 DOI: 10.1039/c9nr05383b] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
A major challenge exists in the preparation of scaffolds for bone regeneration, namely, achieving simultaneously bioactivity, biocompatibility, mechanical performance and simple manufacturing. Here, cellulose nanofibrils (CNF) are introduced for the preparation of scaffolds taking advantage of their biocompatibility and ability to form strong 3D porous networks from aqueous suspensions. CNF are made bioactive for bone formation through a simple and scalable strategy that achieves highly interconnected 3D networks. The resultant materials optimally combine morphological and mechanical features and facilitate hydroxyapatite formation while releasing essential ions for in vivo bone repair. The porosity and roughness of the scaffolds favor several cell functions while the ions act in the expression of genes associated with cell differentiation. Ion release is found critical to enhance the production of the bone morphogenetic protein 2 (BMP-2) from cells within the fractured area, thus accelerating the in vivo bone repair. Systemic biocompatibility indicates no negative effects on vital organs such as the liver and kidneys. The results pave the way towards a facile preparation of advanced, high performance CNF-based scaffolds for bone tissue engineering.
Collapse
Affiliation(s)
- Filipe V Ferreira
- School of Chemical Engineering, University of Campinas (UNICAMP), 13083-970, Campinas-SP, Brazil. and Brazilian Nanotechnology National Laboratory (LNNano), Brazilian Center for Research in Energy and Materials (CNPEM), 13083-970, Campinas-SP, Brazil and Department of Bioproducts and Biosystems, Aalto University School of Chemical Engineering, P.O. Box 16300, 00076, Aalto University, Finland. and Université de Lyon, Ingénierie des Matériaux Polymères CNRS, UMR 5223, INSA Lyon, F-69621 Villeurbanne, France
| | - Lucas P Souza
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), 13083-862, Campinas-SP, Brazil
| | - Thais M M Martins
- Department of Morphology, Institute of Biological Sciences, Federal University of Minas Gerais (UFMG), 31270-901, Belo Horizonte-MG, Brazil
| | - João H Lopes
- Department of Chemistry, Division of Fundamental Sciences (IEF), Technological Institute of Aeronautics (ITA), 12228-900, Sao Jose dos Campos-SP, Brazil
| | - Bruno D Mattos
- Department of Bioproducts and Biosystems, Aalto University School of Chemical Engineering, P.O. Box 16300, 00076, Aalto University, Finland.
| | - Marcos Mariano
- Brazilian Nanotechnology National Laboratory (LNNano), Brazilian Center for Research in Energy and Materials (CNPEM), 13083-970, Campinas-SP, Brazil
| | - Ivanei F Pinheiro
- School of Chemical Engineering, University of Campinas (UNICAMP), 13083-970, Campinas-SP, Brazil. and Brazilian Nanotechnology National Laboratory (LNNano), Brazilian Center for Research in Energy and Materials (CNPEM), 13083-970, Campinas-SP, Brazil
| | - Thalita M Valverde
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais (UFMG), 31270-901, Belo Horizonte-MG, Brazil
| | - Sébastien Livi
- Université de Lyon, Ingénierie des Matériaux Polymères CNRS, UMR 5223, INSA Lyon, F-69621 Villeurbanne, France
| | - José A Camilli
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), 13083-862, Campinas-SP, Brazil
| | - Alfredo M Goes
- Department of Pathology, Institute of Biological Sciences, Federal University of Minas Gerais (UFMG), 31270-901, Belo Horizonte-MG, Brazil
| | - Rubia F Gouveia
- Brazilian Nanotechnology National Laboratory (LNNano), Brazilian Center for Research in Energy and Materials (CNPEM), 13083-970, Campinas-SP, Brazil
| | - Liliane M F Lona
- School of Chemical Engineering, University of Campinas (UNICAMP), 13083-970, Campinas-SP, Brazil.
| | - Orlando J Rojas
- Department of Bioproducts and Biosystems, Aalto University School of Chemical Engineering, P.O. Box 16300, 00076, Aalto University, Finland.
| |
Collapse
|
27
|
Li L, Li J, Zou Q, Zuo Y, Cai B, Li Y. Enhanced bone tissue regeneration of a biomimetic cellular scaffold with co-cultured MSCs-derived osteogenic and angiogenic cells. Cell Prolif 2019; 52:e12658. [PMID: 31297910 PMCID: PMC6797511 DOI: 10.1111/cpr.12658] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 05/09/2019] [Accepted: 05/15/2019] [Indexed: 12/18/2022] Open
Abstract
OBJECTIVES The bone tissue engineering primarily focuses on three-dimensional co-culture systems, which physical and biological properties resemble the cell matrix of actual tissues. The complex dialogue between bone-forming and endothelial cells (ECs) in a tissue-engineered construct will directly regulate angiogenesis and bone regeneration. The purpose of this study was to investigate whether co-culture between osteogenic and angiogenic cells derived by bone mesenchymal stem cells (MSCs) could affect cell activities and new bone formation. MATERIALS AND METHODS Mesenchymal stem cells were dually induced to differentiate into osteogenic cells (OMSCs) and ECs; both cell types were co-cultured at different ratios to investigate their effects and underlying mechanisms through ELISA, RT-qPCR and MTT assays. The selected cell mixture was transplanted onto a nano-hydroxyapatite/polyurethane (n-HA/PU) scaffold to form a cell-scaffold construct that was implanted in the rat femoral condyles. Histology and micro-CT were examined for further verification. RESULTS ELISA and gene expression studies revealed that co-cultured OMSCs/ECs (0.5/1.5) significantly elevated the transcription levels of osteogenic genes such as ALP, Col-I and OCN, as well as transcription factors Msx2, Runx2 and Osterix; it also upregulated angiogenic factors of vascular endothelial growth factor (VEGF) and CD31 when compared with cells cultured alone or in other ratios. The optimized OMSCs/ECs group had more abundant calcium phosphate crystal deposition, further facilitated their bone formation in vivo. CONCLUSIONS The OMSCs/ECs-scaffold constructs at an optimal cell ratio (0.5/1.5) achieved enhanced osteogenic and angiogenic factor expression and biomineralization, which resulted in more effective bone formation.
Collapse
Affiliation(s)
- Limei Li
- Research Center for Nano‐Biomaterials, Analytical & Testing CenterSichuan UniversityChengduChina
- Technology Transfer CenterKunming Medical UniversityKunmingChina
| | - Jidong Li
- Research Center for Nano‐Biomaterials, Analytical & Testing CenterSichuan UniversityChengduChina
| | - Qin Zou
- Research Center for Nano‐Biomaterials, Analytical & Testing CenterSichuan UniversityChengduChina
| | - Yi Zuo
- Research Center for Nano‐Biomaterials, Analytical & Testing CenterSichuan UniversityChengduChina
| | - Bin Cai
- Research Center for Nano‐Biomaterials, Analytical & Testing CenterSichuan UniversityChengduChina
| | - Yubao Li
- Research Center for Nano‐Biomaterials, Analytical & Testing CenterSichuan UniversityChengduChina
| |
Collapse
|
28
|
A facile approach for engineering tissue constructs with vessel-like channels by cell-laden hydrogel fibers. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2019; 101:370-379. [DOI: 10.1016/j.msec.2019.03.094] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 02/20/2019] [Accepted: 03/25/2019] [Indexed: 01/21/2023]
|
29
|
Zhu S, Wang J, Sun Z. Observation of co-culturing cells on porous silk fibroin films. BIOINSPIRED BIOMIMETIC AND NANOBIOMATERIALS 2019. [DOI: 10.1680/jbibn.17.00030] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Porous silk fibroin films (PSFFs) are widely used in skin regeneration. Prevascularization of PSFFs is a successful strategy for enhancing the survival of engineered tissues in vivo. The adhesion and migration of human umbilical vein endothelial cells (HUVECs) and fibroblasts on PSFFs were observed by scanning electron microscopy and confocal laser scanning microscopy after vital staining of the cells. PSFFs could attract a large number of HUVECs and fibroblasts to pores in an interesting alignment and support them to spread well from the outside of pores to the center of PSFFs to form cell layers. PSFFs showed minor structural changes due to less degradation for 12 d culture, and cell layers overlapped from pores to the center in PSFFs. The total DNA assay indicated excellent cell proliferation on PSFFs on days 1 and 6 and no difference between co-culturing HUVECs and mono-HUVECs and monofibroblasts. PSFFs could guide cell migration and arrangement to form pore-centered and vessel-like structures without additional coating of proteins. The authors’ aim was to study the potential prevascularization of porous silk fibroin scaffolds by co-culturing HUVECs and fibroblasts. This model has potential application for angiogenesis in dermis regeneration in the future and provides a suitable microenvironment for the development of capillary-like structures.
Collapse
Affiliation(s)
- Shixin Zhu
- Medical School, Soochow University, Suzhou, China
| | - JiaMin Wang
- Medical School, Soochow University, Suzhou, China
| | - Ziling Sun
- Medical School, Soochow University, Suzhou, China
| |
Collapse
|
30
|
Tang H, Husch JFA, Zhang Y, Jansen JA, Yang F, van den Beucken JJJP. Coculture with monocytes/macrophages modulates osteogenic differentiation of adipose-derived mesenchymal stromal cells on poly(lactic-co-glycolic) acid/polycaprolactone scaffolds. J Tissue Eng Regen Med 2019; 13:785-798. [PMID: 30771241 PMCID: PMC6594112 DOI: 10.1002/term.2826] [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: 10/06/2017] [Revised: 11/17/2018] [Accepted: 02/13/2019] [Indexed: 12/18/2022]
Abstract
The effects of immune cells, in particular macrophages, on the behaviour of mesenchymal stromal cells (MSCs) have recently gained much attention for MSCs‐based tissue‐engineered constructs. This study aimed to evaluate the effect of monocytes/macrophages on the osteogenic differentiation of adipose‐derived mesenchymal stromal cells (ADMSCs) in three‐dimensional (3D) cocultures. For this, we cocultured THP‐1 monocytes, M1 macrophages, or M2 macrophages with ADMSCs on 3D poly(lactic‐co‐glycolic) acid (PLGA)/polycaprolactone (PCL) scaffolds using osteogenic medium for up to 42 days. We found that osteogenic differentiation of ADMSCs was inhibited by monocytes and both macrophage subtypes in 3D scaffolds. Furthermore, coculture of monocytes/macrophages with ADMSCs resulted in downregulated secretion of oncostatin M (OSM) and bone morphogenetic protein 2 (BMP‐2) and inhibited expression of osteogenic markers alkaline phosphatase (ALP), bone sialoprotein (BSP), and runt‐related transcription factor 2 (RUNX2). Compared with both macrophage subtypes, monocytes inhibited osteogenic differentiation of ADMSCs more significantly. These data suggest that the mutual interactions between monocytes/macrophages and ADMSCs negatively affect MSC osteogenic differentiation and thus possibly bone healing capacity, which highlights the importance of the micro‐environment in influencing cell‐based constructs to treat bone defects and the potential to improve their performance by resolving the inflammation ahead of treatment.
Collapse
Affiliation(s)
- Hongbo Tang
- Department of Biomaterials, Radboud Institute for Molecular Life Sciences, Radboudumc, Nijmegen, The Netherlands.,Department of Plastic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Johanna F A Husch
- Department of Biomaterials, Radboud Institute for Molecular Life Sciences, Radboudumc, Nijmegen, The Netherlands
| | - Yang Zhang
- Department of Biomaterials, Radboud Institute for Molecular Life Sciences, Radboudumc, Nijmegen, The Netherlands
| | - John A Jansen
- Department of Biomaterials, Radboud Institute for Molecular Life Sciences, Radboudumc, Nijmegen, The Netherlands
| | - Fang Yang
- Department of Biomaterials, Radboud Institute for Molecular Life Sciences, Radboudumc, Nijmegen, The Netherlands
| | - Jeroen J J P van den Beucken
- Department of Biomaterials, Radboud Institute for Molecular Life Sciences, Radboudumc, Nijmegen, The Netherlands
| |
Collapse
|
31
|
Osteogenic and angiogenic characterization of mandible and femur osteoblasts. J Mol Histol 2019; 50:105-117. [PMID: 30635760 DOI: 10.1007/s10735-019-09810-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Accepted: 01/06/2019] [Indexed: 12/13/2022]
|
32
|
Jiang Y, Li M, Fu X. Biotechnological Management of Angiopathic Wounds: Challenges and Perspectives. INT J LOW EXTR WOUND 2018; 17:214-217. [PMID: 30474446 DOI: 10.1177/1534734618813232] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Angiopathic wound is a wound that develops as a result of a local vascular lesion. Angiogenesis is an important aspect underlying repair, and increased angiogenesis could accelerate and improve the healing outcome. Biotherapy has been used more and more in clinic and brings hope for angiopathic wound treatment, through the rapid recovery of angiogenesis and regulation and correction of the whole wound microenvironment. In this article, we discuss the advantages and disadvantages of various technologies ranging from presentation of angiogenic growth factors, genetic strategies, stem cells, and biomaterials engineering in angiopathic wound treatment.
Collapse
Affiliation(s)
- Yufeng Jiang
- Chinese PLA 306th Hospital, Beijing, People’s Republic of China
- Chinese PLA General Hospital and Chinese PLA Medical College, Beijing, People’s Republic of China
- The Key Laboratory of Wound Repair and Regeneration of PLA, Chinese PLA General Hospital, Beijing, People’s Republic of China
| | - Meirong Li
- Chinese PLA General Hospital and Chinese PLA Medical College, Beijing, People’s Republic of China
- The Key Laboratory of Wound Repair and Regeneration of PLA, Chinese PLA General Hospital, Beijing, People’s Republic of China
| | - Xiaobing Fu
- Chinese PLA General Hospital and Chinese PLA Medical College, Beijing, People’s Republic of China
- The Key Laboratory of Wound Repair and Regeneration of PLA, Chinese PLA General Hospital, Beijing, People’s Republic of China
| |
Collapse
|
33
|
Vácz G, Major B, Gaál D, Petrik L, Horváthy DB, Han W, Holczer T, Simon M, Muir JM, Hornyák I, Lacza Z. Hyperacute serum has markedly better regenerative efficacy than platelet-rich plasma in a human bone oxygen-glucose deprivation model. Regen Med 2018; 13:531-543. [PMID: 30132395 DOI: 10.2217/rme-2017-0141] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
AIM Platelet-rich plasma (PRP) and hyperacute serum (HAS) were compared in a novel human model of ex vivo bone damage induced by oxygen-glucose deprivation (OGD). MATERIALS & METHODS Osteoarthritic subchondral bone pieces were harvested from discarded femoral heads during hip replacement surgery and subjected to transient OGD. RESULTS Proteome profiling revealed that PRP is more angiopoietic, whereas HAS is more antiangiopoietic in composition. However, treatment of OGD-exposed bone with multiple PRP preparations had no effect on cell counts, whereas HAS restored cell proliferation capacity and rescued viable cell number following OGD. CONCLUSION A similar pro-proliferation effect was observed with recombinant growth factors, indicating that HAS may be an alternative agent for enhancing the regeneration of damaged bone cells.
Collapse
Affiliation(s)
- Gabriella Vácz
- Institute of Clinical Experimental Research, Semmelweis University, Tűzoltó u. 37-47, Budapest, Hungary, 1094
| | - Bálint Major
- Polyclinic of the Hospitaller Brothers of St. John of God in Budapest, Orthopaedic Department, Frankel Leo u. 54., Budapest, Hungary, 1023
| | - Dorottya Gaál
- Institute of Clinical Experimental Research, Semmelweis University, Tűzoltó u. 37-47, Budapest, Hungary, 1094
| | - Laura Petrik
- Institute of Clinical Experimental Research, Semmelweis University, Tűzoltó u. 37-47, Budapest, Hungary, 1094
| | - Dénes Balázs Horváthy
- Institute of Clinical Experimental Research, Semmelweis University, Tűzoltó u. 37-47, Budapest, Hungary, 1094
| | - Weiping Han
- Bioimaging Consortium, A-STAR, Singapore, Helios, Biopolis Way 11
| | - Tünde Holczer
- Department of Laboratory Medicine, Semmelweis University, Nagyvárad t. 4, Budapest, Hungary, 1089
| | - Melinda Simon
- Institute of Clinical Experimental Research, Semmelweis University, Tűzoltó u. 37-47, Budapest, Hungary, 1094
| | - Jeffrey M Muir
- Motion Research, 3-35 Stone Church Rd, Suite 215, Ancaster, Ontario, L9K 3S9 Canada
| | - István Hornyák
- OrthoSera GmbH, Dr. Karl-Dorrek-Straße 23-29, 3500 Krems an der Donau, Austria
| | - Zsombor Lacza
- OrthoSera GmbH, Dr. Karl-Dorrek-Straße 23-29, 3500 Krems an der Donau, Austria.,University of Physical Education, Alkotás u. 44, Budapest, Hungary 1123
| |
Collapse
|
34
|
Ritz U, Eberhardt M, Klein A, Frank P, Götz H, Hofmann A, Rommens PM, Jonas U. Photocrosslinked Dextran-Based Hydrogels as Carrier System for the Cells and Cytokines Induce Bone Regeneration in Critical Size Defects in Mice. Gels 2018; 4:E63. [PMID: 30674839 PMCID: PMC6209263 DOI: 10.3390/gels4030063] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 06/30/2018] [Accepted: 07/03/2018] [Indexed: 01/07/2023] Open
Abstract
Modified biomaterials have for years been the focus of research into establishing new bone substitutes. In our preceding in vitro study employing different cell cultures, we developed chemically and mechanically characterized hydrogels based on photocrosslinkable dextran derivatives and demonstrated their cytocompatibility and their beneficial effects on the proliferation of osteoblasts and endothelial cells. In the present in vivo study, we investigate photocrosslinked dextran-based hydrogels in critical size defects in mice to evaluate their potential as carrier systems for cells or for a specific angiogenesis enhancing cytokine to induce bone formation. We could demonstrate that, with optimized laboratory practice, the endotoxin content of hydrogels could be reduced below the Food and Drug Administration (FDA)-limit. Dextran-based hydrogels were either loaded with a monoculture of endothelial cells or a co-culture of human osteoblasts with endothelial cells, or with stromal-derived-growth factor (SDF-1). Scaffolds were implanted into a calvarial defect of critical size in mice and their impact on bone formation was assessed by µCt-analyses, histology and immunohistology. Our study demonstrates that promotion of angiogenesis either by SDF-1 or a monoculture of endothelial cells induces bone regeneration at a physiological level. These in vivo results indicate the potential of dextran-based hydrogel composites in bone regeneration to deliver cells and cytokines to the defect site.
Collapse
Affiliation(s)
- Ulrike Ritz
- Biomatics Group, Department of Orthopaedics and Traumatology, University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany.
| | - Marc Eberhardt
- Biomatics Group, Department of Orthopaedics and Traumatology, University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany.
| | - Anja Klein
- Biomatics Group, Department of Orthopaedics and Traumatology, University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany.
| | - Petra Frank
- Macromolecular Chemistry, Department Chemistry Biology, University of Siegen, 57076 Siegen, Germany.
| | - Hermann Götz
- Biomatics Group, Platform Biomaterials, University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany.
| | - Alexander Hofmann
- Biomatics Group, Department of Orthopaedics and Traumatology, University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany.
| | - Pol Maria Rommens
- Biomatics Group, Department of Orthopaedics and Traumatology, University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany.
| | - Ulrich Jonas
- Macromolecular Chemistry, Department Chemistry Biology, University of Siegen, 57076 Siegen, Germany.
| |
Collapse
|
35
|
Abstract
Creating an optimal microenvironment that supports angiogenesis, cell-cell cross talk, cell migration, and differentiation is crucial for pulp/dentin regeneration. It was shown that dental stem cells being seeded onto a scaffold and transplanted in vivo could give rise to a new tissue similar to that of the native pulp. However, the unique structure of the tooth with a pulp space encased within hard dentin allows only a single blood supply from a small apical opening located at the apex of the root canals. Therefore, a further strategy that can address this limitation such as the incorporation of endothelial/endothelial progenitor cells or cells with high angiogenic potential into the transplant is required so that the added cells can contribute to the vascularization within the implant. However, the placement of 2 or more different cell types inside 3-dimensional porous scaffolds is technologically challenging. In contrast to the conventional scaffolding approach, self-assembly of monodispersed cells into 3-dimensional tissue mimics permits true physiological interactions between and among different types of cells without any influence from a secondary material. In this review, we discuss potential strategies that can be used in vasculature engineering in dental pulp regeneration with a specific emphasis on combining prevascularization and scaffold-based or scaffold-free approaches.
Collapse
Affiliation(s)
| | - Chengfei Zhang
- Endodontology, Faculty of Dentistry, The University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
36
|
Yuan Q, Arkudas A, Horch RE, Hammon M, Bleiziffer O, Uder M, Seuss H. Vascularization of the Arteriovenous Loop in a Rat Isolation Chamber Model—Quantification of Hypoxia and Evaluation of Its Effects. Tissue Eng Part A 2018; 24:719-728. [DOI: 10.1089/ten.tea.2017.0262] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Affiliation(s)
- Quan Yuan
- Department of Plastic and Hand Surgery, Laboratory for Tissue Engineering and Regenerative Medicine, University Hospital Erlangen, Friedrich Alexander University, Erlangen-Nuernberg (FAU), Erlangen, Germany
- Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Andreas Arkudas
- Department of Plastic and Hand Surgery, Laboratory for Tissue Engineering and Regenerative Medicine, University Hospital Erlangen, Friedrich Alexander University, Erlangen-Nuernberg (FAU), Erlangen, Germany
| | - Raymund E. Horch
- Department of Plastic and Hand Surgery, Laboratory for Tissue Engineering and Regenerative Medicine, University Hospital Erlangen, Friedrich Alexander University, Erlangen-Nuernberg (FAU), Erlangen, Germany
| | - Matthias Hammon
- Department of Radiology, University Hospital Erlangen, Friedrich Alexander University, Erlangen-Nuernberg (FAU), Erlangen, Germany
| | - Oliver Bleiziffer
- Department of Plastic and Hand Surgery, Laboratory for Tissue Engineering and Regenerative Medicine, University Hospital Erlangen, Friedrich Alexander University, Erlangen-Nuernberg (FAU), Erlangen, Germany
- Department of Plastic and Hand Surgery, Inselspital Bern, Universität Bern, Bern, Switzerland
| | - Michael Uder
- Department of Radiology, University Hospital Erlangen, Friedrich Alexander University, Erlangen-Nuernberg (FAU), Erlangen, Germany
| | - Hannes Seuss
- Department of Radiology, University Hospital Erlangen, Friedrich Alexander University, Erlangen-Nuernberg (FAU), Erlangen, Germany
| |
Collapse
|
37
|
Pirosa A, Gottardi R, Alexander PG, Tuan RS. Engineering in-vitro stem cell-based vascularized bone models for drug screening and predictive toxicology. Stem Cell Res Ther 2018; 9:112. [PMID: 29678192 PMCID: PMC5910611 DOI: 10.1186/s13287-018-0847-8] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The production of veritable in-vitro models of bone tissue is essential to understand the biology of bone and its surrounding environment, to analyze the pathogenesis of bone diseases (e.g., osteoporosis, osteoarthritis, osteomyelitis, etc.), to develop effective therapeutic drug screening, and to test potential therapeutic strategies. Dysregulated interactions between vasculature and bone cells are often related to the aforementioned pathologies, underscoring the need for a bone model that contains engineered vasculature. Due to ethical restraints and limited prediction power of animal models, human stem cell-based tissue engineering has gained increasing relevance as a candidate approach to overcome the limitations of animals and to serve as preclinical models for drug testing. Since bone is a highly vascularized tissue, the concomitant development of vasculature and mineralized matrix requires a synergistic interaction between osteogenic and endothelial precursors. A number of experimental approaches have been used to achieve this goal, such as the combination of angiogenic factors and three-dimensional scaffolds, prevascularization strategies, and coculture systems. In this review, we present an overview of the current models and approaches to generate in-vitro stem cell-based vascularized bone, with emphasis on the main challenges of vasculature engineering. These challenges are related to the choice of biomaterials, scaffold fabrication techniques, and cells, as well as the type of culturing conditions required, and specifically the application of dynamic culture systems using bioreactors.
Collapse
Affiliation(s)
- Alessandro Pirosa
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, 450 Technology Drive, Pittsburgh, PA 15219 USA
| | - Riccardo Gottardi
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, 450 Technology Drive, Pittsburgh, PA 15219 USA
- Ri.MED Foundation, Via Bandiera 11, Palermo, 90133 Italy
| | - Peter G. Alexander
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, 450 Technology Drive, Pittsburgh, PA 15219 USA
| | - Rocky S. Tuan
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, 450 Technology Drive, Pittsburgh, PA 15219 USA
| |
Collapse
|
38
|
Klein A, Baranowski A, Ritz U, Götz H, Heinemann S, Mattyasovszky S, Rommens PM, Hofmann A. Effect of bone sialoprotein coated three-dimensional printed calcium phosphate scaffolds on primary human osteoblasts. J Biomed Mater Res B Appl Biomater 2018; 106:2565-2575. [PMID: 29316208 DOI: 10.1002/jbm.b.34073] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 11/29/2017] [Accepted: 12/22/2017] [Indexed: 01/14/2023]
Abstract
The combination of the two techniques of rapid prototyping 3D-plotting and bioactive surface functionalization is presented, with emphasis on the in vitro effect of Bone Sialoprotein (BSP) on primary human osteoblasts (hOBs). Our primary objective was to demonstrate the BSP influence on the expression of distinctive osteoblast markers in hOBs. Secondary objectives included examinations of the scaffolds' surface and the stability of BSP-coating as well as investigations of cell viability and proliferation. 3D-plotted calcium phosphate cement (CPC) scaffolds were coated with BSP via physisorption. hOBs were seeded on the coated scaffolds, followed by cell viability measurements, gene expression analysis and visualization. Physisorption is an effective method for BSP-coating. Coating with higher BSP concentrations leads to enhanced BSP release. Two BSP concentrations (50 and 200 μg/mL) were examined in this study. The lower BSP concentration (50 µg/mL) decreased ALP and SPARC expression, whereas the higher BSP concentration (200 μg/mL) did not change gene marker expression. Enhanced cell viability was observed on BSP-coated scaffolds on day 3. hOBs developed a polygonal shape and connected in an intercellular network under BSP influence. Quantitative cell morphology analyses demonstrated for BSP-coated CPCs an enhanced cell area and reduced circularity. The strength of the above-mentioned effects of BSP-coated scaffolds in vivo is unknown, and future work is focusing on bone ingrowth and vascularization in vivo. © 2018 Wiley Periodicals, Inc. J Biomed Mater Res Part B: Appl Biomater, 106B: 2565-2575, 2018.
Collapse
Affiliation(s)
- Anja Klein
- Department of Orthopaedics and Traumatology, Biomatics Group, University Medical Center, Johannes Gutenberg University, Mainz, Germany
| | - Andreas Baranowski
- Department of Orthopaedics and Traumatology, Biomatics Group, University Medical Center, Johannes Gutenberg University, Mainz, Germany
| | - Ulrike Ritz
- Department of Orthopaedics and Traumatology, Biomatics Group, University Medical Center, Johannes Gutenberg University, Mainz, Germany
| | - Hermann Götz
- Platform for Biomaterial Research, Biomatics Group, University Medical Center, Johannes Gutenberg University, Mainz, Germany
| | | | - Stefan Mattyasovszky
- Department of Orthopaedics and Traumatology, Biomatics Group, University Medical Center, Johannes Gutenberg University, Mainz, Germany
| | - Pol M Rommens
- Department of Orthopaedics and Traumatology, Biomatics Group, University Medical Center, Johannes Gutenberg University, Mainz, Germany
| | - Alexander Hofmann
- Department of Orthopaedics and Traumatology, Biomatics Group, University Medical Center, Johannes Gutenberg University, Mainz, Germany
| |
Collapse
|
39
|
Yan Z, Yin H, Nerlich M, Pfeifer CG, Docheva D. Boosting tendon repair: interplay of cells, growth factors and scaffold-free and gel-based carriers. J Exp Orthop 2018; 5:1. [PMID: 29330711 PMCID: PMC5768579 DOI: 10.1186/s40634-017-0117-1] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Accepted: 12/20/2017] [Indexed: 12/21/2022] Open
Abstract
Background Tendons are dense connective tissues and critical components for the integrity and function of the musculoskeletal system. Tendons connect bone to muscle and transmit forces on which locomotion entirely depends. Due to trauma, overuse and age-related degeneration, many people suffer from acute or chronic tendon injuries. Owing to their hypovascularity and hypocellularity, tendinopathies remain a substantial challenge for both clinicians and researchers. Surgical treatment includes suture or transplantation of autograft, allograft or xenograft, and these serve as the most common technique for rescuing tendon injuries. However, the therapeutic efficacies are limited by drawbacks including inevitable donor site morbidity, poor graft integration, adhesion formations and high rates of recurrent tearing. This review summarizes the literature of the past 10 y concerning scaffold-free and gel-based approaches for treating tendon injuries, with emphasis on specific advantages of such modes of application, as well as the obtained results regarding in vitro and in vivo tenogenesis. Results The search was focused on publications released after 2006 and 83 articles have been analysed. The main results are summarizing and discussing the clear advantages of scaffold-free and hydrogels carriers that can be functionalized with cells alone or in combination with growth factors. Conclusion The improved understanding of tissue resident adult stem cells has made a significant progress in recent years as well as strategies to steer their fate toward tendon lineage, with the help of growth factors, have been identified. The field of tendon tissue engineering is exploring diverse models spanning from hard scaffolds to gel-based and scaffold-free approaches seeking easier cell delivery and integration in the site of injury. Still, the field needs to consider a multifactorial approach that is based on the combination and fine-tuning of chemical and biomechanical stimuli. Taken together, tendon tissue engineering has now excellent foundations and enters the period of precision and translation to models with clinical relevance on which better treatment options of tendon injuries can be shaped up.
Collapse
Affiliation(s)
- Zexing Yan
- Laboratory of Experimental Trauma Surgery, Department of Trauma Surgery, University Regensburg Medical Centre, Franz-Josef-Strauss-Allee 11, 93053, Regensburg, Germany
| | - Heyong Yin
- Laboratory of Experimental Trauma Surgery, Department of Trauma Surgery, University Regensburg Medical Centre, Franz-Josef-Strauss-Allee 11, 93053, Regensburg, Germany
| | - Michael Nerlich
- Laboratory of Experimental Trauma Surgery, Department of Trauma Surgery, University Regensburg Medical Centre, Franz-Josef-Strauss-Allee 11, 93053, Regensburg, Germany
| | - Christian G Pfeifer
- Laboratory of Experimental Trauma Surgery, Department of Trauma Surgery, University Regensburg Medical Centre, Franz-Josef-Strauss-Allee 11, 93053, Regensburg, Germany
| | - Denitsa Docheva
- Laboratory of Experimental Trauma Surgery, Department of Trauma Surgery, University Regensburg Medical Centre, Franz-Josef-Strauss-Allee 11, 93053, Regensburg, Germany. .,Director of Experimental Trauma Surgery, Department of Trauma Surgery, University Regensburg Medical Centre, Franz-Josef-Strauss-Allee 11, 93053, Regensburg, Germany.
| |
Collapse
|
40
|
Zhang C, Hu K, Liu X, Reynolds MA, Bao C, Wang P, Zhao L, Xu HH. Novel hiPSC-based tri-culture for pre-vascularization of calcium phosphate scaffold to enhance bone and vessel formation. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2017. [DOI: 10.1016/j.msec.2017.05.035] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
41
|
Manipulation of Human Primary Endothelial Cell and Osteoblast Coculture Ratios to Augment Vasculogenesis and Mineralization. Ann Plast Surg 2017; 77:122-8. [PMID: 25144419 DOI: 10.1097/sap.0000000000000318] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Tissue-engineering scaffolds are often seeded with a single type of cell, but there has been more focus on cocultures to improve angiogenesis and bone formation for craniofacial applications. Investigation of bone-derived osteoblasts (OBs) is important because of the use of bone grafts and migration of OBs from native bone into constructs in vivo and therefore, their contribution to bone formation in vivo. The limitation of primary OBs has been their inability to mineralize without osteogenic factors in vitro. Through coculture of OBs and endothelial cells (ECs) and manipulation of the coculture ratio, mineralization can be achieved without osteogenic media or additional growth factors, thus enhancing their utility for tissue-engineering applications. An optimal ratio of EC/OB for vasculogenesis and mineralization has not been determined for human primary cells. Human umbilical vein ECs were cultured with normal human primary OBs in different EC/OB ratios, namely, 10:1, 5:1, 1:1, 1:5, and 1:10 with EC and OB monocultures as controls. The number of vasculogenic networks in a collagen matrix was highest in ratios of 5:1 and 1:1. ECs lined up and formed capillary-like networks by day 10, which was not seen in the other groups. On polystyrene, cells were cocultured with ECs and OBs in direct contact (direct coculture) or separated by a transwell membrane (indirect coculture). At day 21, Alizarin Red staining showed mineralization on the 1:5 and 1:10 direct coculture ratios, with 1:5 having more mineralization nodules present than 1:10. No mineralization was seen in other direct coculture ratios or in any of the indirect coculture ratios. Alkaline phosphatase secretion was highest in the 1:5 direct coculture group. Vascular endothelial growth factor secretion from OBs was present in the 1:5 and 1:10 direct coculture ratios at all time points and inhibited after day 1 in other coculture groups. To improve vasculogenesis, cocultures of primary human ECs and OBs in ratios of 5:1 should be used, but to improve bone formation, the 1:5 direct coculture ratio results in most mineralization.
Collapse
|
42
|
Lee S, Lee K, Kim SH, Jung Y. Enhanced Cartilaginous Tissue Formation with a Cell Aggregate-Fibrin-Polymer Scaffold Complex. Polymers (Basel) 2017; 9:E348. [PMID: 30971025 PMCID: PMC6418534 DOI: 10.3390/polym9080348] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Revised: 08/03/2017] [Accepted: 08/03/2017] [Indexed: 12/23/2022] Open
Abstract
Cell density is one of the factors required in the preparation of engineered cartilage from mesenchymal stem cells (MSCs). Additionally, it is well known for having a significant role in chemical and physical stimulations when stem cells undergo chondrogenic differentiation. Here, we developed an engineered cartilage with a cell aggregate-hydrogel-polymer scaffold complex capable of inducing the effective regeneration of cartilage tissue similar to natural cartilage while retaining a high mechanical strength, flexibility, and morphology. Cell aggregates were generated by the hanging drop method with rabbit bone marrow stromal cells (BMSCs), and poly (lactide-co-caprolactone) (PLCL) scaffolds were fabricated with 78.3 ± 5.3% porosity and a 300⁻500 μm pore size with a gel-pressing method. We prepared the cell aggregate-fibrin-poly (lactide-co-caprolactone) (PLCL) scaffold complex, in which the cell aggregates were evenly dispersed in the fibrin, and they were immobilized onto the surface of the polymer scaffold while filling up the pores. To examine the chondrogenic differentiation of seeded BMSCs and the formation of chondral extracellular matrix onto the complexes, they were cultured in vitro or subcutaneously implanted into nude mice for up to eight weeks. The results of the in vitro and in vivo studies revealed that the accumulation of the chondral extracellular matrices was increased on the cell aggregate-fibrin-PLCL scaffold complexes (CAPs) compared to the single cell-fibrin-PLCL scaffold complexes (SCPs). Additionally, we examined whether the mature and well-developed cartilaginous tissues and lacunae structures typical of mature cartilage were evenly distributed in the CAPs. Consequently, the cell aggregates in the hybrid scaffolds of fibrin gels and elastic PLCL scaffolds can induce themselves to differentiate into chondrocytes, maintain their phenotypes, enhance glycosaminoglycan (GAG) production, and improve the quality of cartilaginous tissue formed in vitro and in vivo.
Collapse
Affiliation(s)
- Soojin Lee
- Biomaterials Research Center, Korea Institute of Science and Technology, 5, Hwarang-ro 14-Gil, Seoungbuk-gu, Seoul 02792, Korea.
- Program in Nanoscience and Technology, Graduate School of Convergence Science and Technology, Seoul National University, Seoul 08826, Korea.
| | - Kangwon Lee
- Program in Nanoscience and Technology, Graduate School of Convergence Science and Technology, Seoul National University, Seoul 08826, Korea.
- Advanced Institutes of Convergence Technology, Gyeonggi-do 16229, Korea.
| | - Soo Hyun Kim
- Biomaterials Research Center, Korea Institute of Science and Technology, 5, Hwarang-ro 14-Gil, Seoungbuk-gu, Seoul 02792, Korea.
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Korea.
| | - Youngmee Jung
- Biomaterials Research Center, Korea Institute of Science and Technology, 5, Hwarang-ro 14-Gil, Seoungbuk-gu, Seoul 02792, Korea.
- Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology, Seoul 02792, Korea.
| |
Collapse
|
43
|
Um Min Allah N, Berahim Z, Ahmad A, Kannan TP. Biological Interaction Between Human Gingival Fibroblasts and Vascular Endothelial Cells for Angiogenesis: A Co-culture Perspective. Tissue Eng Regen Med 2017; 14:495-505. [PMID: 30603504 DOI: 10.1007/s13770-017-0065-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Revised: 05/18/2017] [Accepted: 06/08/2017] [Indexed: 12/13/2022] Open
Abstract
Advancement in cell culture protocols, multidisciplinary research approach, and the need of clinical implication to reconstruct damaged or diseased tissues has led to the establishment of three-dimensional (3D) test systems for regeneration and repair. Regenerative therapies, including dental tissue engineering, have been pursued as a new prospect to repair and rebuild the diseased/lost oral tissues. Interactions between the different cell types, growth factors, and extracellular matrix components involved in angiogenesis are vital in the mechanisms of new vessel formation for tissue regeneration. In vitro pre-vascularization is one of the leading scopes in the tissue-engineering field. Vascularization strategies that are associated with co-culture systems have proved that there is communication between different cell types with mutual beneficial effects in vascularization and tissue regeneration in two-dimensional or 3D cultures. Endothelial cells with different cell populations, including osteoblasts, smooth muscle cells, and fibroblasts in a co-culture have shown their ability to advocate pre-vascularization. In this review, a co-culture perspective of human gingival fibroblasts and vascular endothelial cells is discussed with the main focus on vascularization and future perspective of this model in regeneration and repair.
Collapse
Affiliation(s)
- Nasar Um Min Allah
- 1School of Dental Sciences, Universiti Sains Malaysia, 16150 Kubang Kerian, Kelantan Malaysia
| | - Zurairah Berahim
- 1School of Dental Sciences, Universiti Sains Malaysia, 16150 Kubang Kerian, Kelantan Malaysia
| | - Azlina Ahmad
- 1School of Dental Sciences, Universiti Sains Malaysia, 16150 Kubang Kerian, Kelantan Malaysia
| | - Thirumulu Ponnuraj Kannan
- 1School of Dental Sciences, Universiti Sains Malaysia, 16150 Kubang Kerian, Kelantan Malaysia
- 2Human Genome Centre, School of Medical Sciences, Universiti Sains Malaysia, 16150 Kubang Kerian, Kelantan Malaysia
| |
Collapse
|
44
|
Marzec M, Kucińska-Lipka J, Kalaszczyńska I, Janik H. Development of polyurethanes for bone repair. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2017; 80:736-747. [PMID: 28866223 DOI: 10.1016/j.msec.2017.07.047] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Revised: 01/23/2017] [Accepted: 07/29/2017] [Indexed: 12/12/2022]
Abstract
The purpose of this paper is to review recent developments on polyurethanes aimed at the design, synthesis, modifications, and biological properties in the field of bone tissue engineering. Different polyurethane systems are presented and discussed in terms of biodegradation, biocompatibility and bioactivity. A comprehensive discussion is provided of the influence of hard to soft segments ratio, catalysts, stiffness and hydrophilicity of polyurethanes. Interaction with various cells, behavior in vivo and current strategies in enhancing bioactivity of polyurethanes are described. The discussion on the incorporation of biomolecules and growth factors, surface modifications, and obtaining polyurethane-ceramics composites strategies is held. The main emphasis is placed on the progress of polyurethane applications in bone regeneration, including bone void fillers, shape memory scaffolds, and drug carrier.
Collapse
Affiliation(s)
- M Marzec
- Department of Polymer Technology, Faculty of Chemistry, Gdansk University of Technology, Narutowicza 11/12, 80-233 Gdansk, Poland
| | - J Kucińska-Lipka
- Department of Polymer Technology, Faculty of Chemistry, Gdansk University of Technology, Narutowicza 11/12, 80-233 Gdansk, Poland.
| | - I Kalaszczyńska
- Department of Histology and Embryology, Center for Biostructure Research, Medical University of Warsaw, Chalubinskiego 5, 02-004 Warsaw, Poland; Centre for Preclinical Research and Technology, Banacha 1b, 02-097 Warsaw, Poland
| | - H Janik
- Department of Polymer Technology, Faculty of Chemistry, Gdansk University of Technology, Narutowicza 11/12, 80-233 Gdansk, Poland
| |
Collapse
|
45
|
Hertweck J, Ritz U, Götz H, Schottel PC, Rommens PM, Hofmann A. CD34 + cells seeded in collagen scaffolds promote bone formation in a mouse calvarial defect model. J Biomed Mater Res B Appl Biomater 2017; 106:1505-1516. [PMID: 28730696 DOI: 10.1002/jbm.b.33956] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Revised: 05/29/2017] [Accepted: 07/04/2017] [Indexed: 11/10/2022]
Abstract
Bone tissue engineering (BTE) holds promise for managing the clinical problem of large bone defects. However, clinical adoption of BTE is limited due to limited vascularization of constructs, which could be circumvented by pre-cultivation of osteogenic and endothelial derived cells in natural-based polymer scaffolds. However, until now not many studies compared the effect of mono- and cocultures pre-seeded in collagen before implantation. We utilized a mouse calvarial defect model and compared five groups of collagen scaffolds: a negative control of a collagen scaffold alone, a positive control treated with BMP-7, monocultures of either human osteoblasts (hOBs) or CD34+ cells, and a coculture of hOB and CD34+ cells. Each pre-seeded collagen scaffold was implanted in mice. After 6 weeks mice were sacrificed and their skulls prepared for volumetric and histologic analysis. We found that a monoculture of CD34+ cells and a coculture of hOB and CD34+ cells pre-cultured in the collagen scaffold increased bone regeneration to a similar extend. In these groups, greater amounts of new bone were found compared with hOB monocultures. Interestingly, monoculture of CD34+ cells demonstrated better fracture healing than monoculture of hOBs, emphasizing the possible role of angiogenesis. Our results are promising regarding a cellular based collagen BTE construct, but more work is needed to understand the complex interaction between the osteogenic and endothelial cells. © 2017 Wiley Periodicals, Inc. J Biomed Mater Res Part B: Appl Biomater, 106B: 1505-1516, 2018.
Collapse
Affiliation(s)
- Jens Hertweck
- Department of Orthopaedics and Traumatology, University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Ulrike Ritz
- Department of Orthopaedics and Traumatology, University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Hermann Götz
- Platform for Biomaterial Research, Biomatics, University Medical Centre, Johannes Gutenberg University, Mainz, Germany
| | - Patrick C Schottel
- Department of Orthopedics and Rehabilitation, University of Vermont Medical Center, Burlington, Vermont
| | - Pol Maria Rommens
- Department of Orthopaedics and Traumatology, University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Alexander Hofmann
- Department of Orthopaedics and Traumatology, University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany
| |
Collapse
|
46
|
Tang H, Zhang Y, Jansen JA, van den Beucken JJJP. Effect of monocytes/macrophages on the osteogenic differentiation of adipose-derived mesenchymal stromal cells in 3D co-culture spheroids. Tissue Cell 2017; 49:461-469. [PMID: 28684045 DOI: 10.1016/j.tice.2017.06.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Revised: 06/02/2017] [Accepted: 06/06/2017] [Indexed: 02/08/2023]
Abstract
This study aimed to investigate the distinctive roles of the monocytes and macrophages on osteogenic differentiation of adipose-derived mesenchymal stromal cells (ADMSCs) in 3D spheroid co-cultures. We hypothesized that monocytes or macrophages (subtypes pro-inflammatory M1 and pro-wound healing M2) would affect the osteogenic differentiation of ADMSCs in 3D spheroids and that cell-cell interactions between monocytes/macrophages and ADMSCs play an important role in the osteogenic differentiation process of ADMSCs. The obtained results indicated that the osteogenic differentiation of ADMSCs was inhibited by monocytes and both macrophage subtypes in 3D spheroids. Monocytes and M2 macrophages had a stronger inhibiting effect than M1 macrophages. Cell-cell interactions mediated by N-cadherin likely played a role in the inhibiting effect of monocytes/macrophages on the osteogenic differentiation of ADMSCs.
Collapse
Affiliation(s)
- Hongbo Tang
- Department of Biomaterials, Radboudumc, Nijmegen, the Netherlands; Department of Plastic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yang Zhang
- Department of Biomaterials, Radboudumc, Nijmegen, the Netherlands
| | - John A Jansen
- Department of Biomaterials, Radboudumc, Nijmegen, the Netherlands
| | | |
Collapse
|
47
|
Shi Y, Wang F, Tiwari S, Yesilbas M, Steubesand N, Weitkamp JT, Klüter T, Lippross S, Eglin D, Seekamp A, Fuchs S. Role of myeloid early endothelial progenitor cells in bone formation and osteoclast differentiation in tissue construct based on hydroxyapatite poly(ester-urethane) scaffolds. J Orthop Res 2016; 34:1922-1932. [PMID: 26945676 DOI: 10.1002/jor.23222] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Accepted: 02/23/2016] [Indexed: 02/04/2023]
Abstract
Engineering of a vascularized bone construct is a highly challenging task which needs to take into account the impact of different components on the bone regeneration process. Bone repair influencing factors in such constructs range from the material properties and scaffold design, to the interaction of different cell types contributing to bone formation and remodeling or neovascularization, respectively. In this context, early endothelial progenitor cells (EPC), mononuclear cells isolated from the peripheral blood, express the endothelial marker CD31 but also a series of myeloid markers and have been shown to support the formation of vessel-like structures. These cells are also characterized by a highly adaptable phenotype influenced by other cells creating an instructive niche. The present study was designed to investigate the impact of EPC on bone formation or remodeling using a co-culture system of outgrowth endothelial cells, mature endothelial cells isolated from the peripheral blood cell cultures, and mesenchymal stem cells grown on hydroxyapatite poly(ester-urethane) scaffolds. The formation of vessel-like structures in these constructs was shown by CLSM and immunohistochemistry and further evaluated by real time RT-PCR. Osteogenic differentiation in these constructs was investigated by von Kossa, Alizarin Red, and real time PCR. Data indicated that osteogenic differentiation occurred within the constructs after 14 days of culture but without a direct influence by EPC in this process. Finally, although we observed a series of osteoclast related makers in the constructs when EPC were included, no indications for an increased osteoclast-like activity, which might lead to increased bone resorption, were observed. © 2016 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 34:1922-1932, 2016.
Collapse
Affiliation(s)
- Yang Shi
- Experimental Trauma Surgery, Arnold-Heller-Straße 3, University Medical Center Schleswig-Holstein, UKSH, 24105, Kiel, Germany
| | - Fanlu Wang
- Experimental Trauma Surgery, Arnold-Heller-Straße 3, University Medical Center Schleswig-Holstein, UKSH, 24105, Kiel, Germany
| | - Sanjay Tiwari
- Molecular Imaging North Competence Center (MOINCC), Kiel, Germany
| | - Meran Yesilbas
- Experimental Trauma Surgery, Arnold-Heller-Straße 3, University Medical Center Schleswig-Holstein, UKSH, 24105, Kiel, Germany
| | - Nadine Steubesand
- Experimental Trauma Surgery, Arnold-Heller-Straße 3, University Medical Center Schleswig-Holstein, UKSH, 24105, Kiel, Germany
| | - Jan-Tobias Weitkamp
- Experimental Trauma Surgery, Arnold-Heller-Straße 3, University Medical Center Schleswig-Holstein, UKSH, 24105, Kiel, Germany
| | - Tim Klüter
- Experimental Trauma Surgery, Arnold-Heller-Straße 3, University Medical Center Schleswig-Holstein, UKSH, 24105, Kiel, Germany
| | - Sebastian Lippross
- Experimental Trauma Surgery, Arnold-Heller-Straße 3, University Medical Center Schleswig-Holstein, UKSH, 24105, Kiel, Germany
| | - David Eglin
- AO Research Institute Davos, Davos, Switzerland
| | - Andreas Seekamp
- Experimental Trauma Surgery, Arnold-Heller-Straße 3, University Medical Center Schleswig-Holstein, UKSH, 24105, Kiel, Germany
| | - Sabine Fuchs
- Experimental Trauma Surgery, Arnold-Heller-Straße 3, University Medical Center Schleswig-Holstein, UKSH, 24105, Kiel, Germany
| |
Collapse
|
48
|
Influence of co-culture on osteogenesis and angiogenesis of bone marrow mesenchymal stem cells and aortic endothelial cells. Microvasc Res 2016; 108:1-9. [DOI: 10.1016/j.mvr.2016.06.005] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Revised: 05/11/2016] [Accepted: 06/27/2016] [Indexed: 11/20/2022]
|
49
|
Freeman FE, Stevens HY, Owens P, Guldberg RE, McNamara LM. Osteogenic Differentiation of Mesenchymal Stem Cells by Mimicking the Cellular Niche of the Endochondral Template. Tissue Eng Part A 2016; 22:1176-1190. [PMID: 27604384 DOI: 10.1089/ten.tea.2015.0339] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
In vitro bone regeneration strategies that prime mesenchymal stem cells (MSCs) with chondrogenic factors, to mimic aspects of the endochondral ossification process, have been shown to promote mineralization and vascularization by MSCs both in vitro and when implanted in vivo. However, these approaches required the use of osteogenic supplements, namely dexamethasone, ascorbic acid, and β-glycerophosphate, none of which are endogenous mediators of bone formation in vivo. Rather MSCs, endothelial progenitor cells, and chondrocytes all reside in proximity within the cartilage template and might paracrineally regulate osteogenic differentiation. Thus, this study tests the hypothesis that an in vitro bone regeneration approach that mimics the cellular niche existing during endochondral ossification, through coculture of MSCs, endothelial cells, and chondrocytes, will obviate the need for extraneous osteogenic supplements and provide an alternative strategy to elicit osteogenic differentiation of MSCs and mineral production. The specific objectives of this study were to (1) mimic the cellular niche existing during endochondral ossification and (2) investigate whether osteogenic differentiation could be induced without the use of any external growth factors. To test the hypothesis, we evaluated the mineralization and vessel formation potential of (a) a novel methodology involving both chondrogenic priming and the coculture of human umbilical vein endothelial cells (HUVECs) and MSCs compared with (b) chondrogenic priming of MSCs alone, (c) addition of HUVECs to chondrogenically primed MSC aggregates, (d-f) the same experimental groups cultured in the presence of osteogenic supplements and (g) a noncoculture group cultured in the presence of osteogenic growth factors alone. Biochemical (DNA, alkaline phosphatase [ALP], calcium, CD31+, vascular endothelial growth factor [VEGF]), histological (alcian blue, alizarin red), and immunohistological (CD31+) analyses were conducted to investigate osteogenic differentiation and vascularization at various time points (1, 2, and 3 weeks). The coculture methodology enhanced both osteogenesis and vasculogenesis compared with osteogenic differentiation alone, whereas osteogenic supplements inhibited the osteogenesis and vascularization (ALP, calcium, and VEGF) induced through coculture alone. Taken together, these results suggest that chondrogenic and vascular priming can obviate the need for osteogenic supplements to induce osteogenesis of human MSCs in vitro, while allowing for the formation of rudimentary vessels.
Collapse
Affiliation(s)
- Fiona E Freeman
- 1 Biomedical Engineering, Centre for Biomechanics Research (BMEC), National University of Ireland Galway , Galway, Ireland
| | - Hazel Y Stevens
- 2 George W. Woodruff School of Mechanical Engineering, Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology , Atlanta, Georgia
| | - Peter Owens
- 3 Centre for Microscopy and Imaging, National University of Ireland , Galway, Galway, Ireland
| | - Robert E Guldberg
- 2 George W. Woodruff School of Mechanical Engineering, Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology , Atlanta, Georgia
| | - Laoise M McNamara
- 1 Biomedical Engineering, Centre for Biomechanics Research (BMEC), National University of Ireland Galway , Galway, Ireland
| |
Collapse
|
50
|
Human Bone Xenografts: from Preclinical Testing for Regenerative Medicine to Modeling of Diseases. ACTA ACUST UNITED AC 2016. [DOI: 10.1007/s40610-016-0044-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|