1
|
El Hajj S, Ntaté MB, Breton C, Siadous R, Aid R, Dupuy M, Letourneur D, Amédée J, Duval H, David B. Bone Spheroid Development Under Flow Conditions with Mesenchymal Stem Cells and Human Umbilical Vein Endothelial Cells in a 3D Porous Hydrogel Supplemented with Hydroxyapatite. Gels 2024; 10:666. [PMID: 39451319 PMCID: PMC11506954 DOI: 10.3390/gels10100666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 10/07/2024] [Accepted: 10/11/2024] [Indexed: 10/26/2024] Open
Abstract
Understanding the niche interactions between blood and bone through the in vitro co-culture of osteo-competent cells and endothelial cells is a key factor in unraveling therapeutic potentials in bone regeneration. This can be additionally supported by employing numerical simulation techniques to assess local physical factors, such as oxygen concentration, and mechanical stimuli, such as shear stress, that can mediate cellular communication. In this study, we developed a Mesenchymal Stem Cell line (MSC) and a Human Umbilical Vein Endothelial Cell line (HUVEC), which were co-cultured under flow conditions in a three-dimensional, porous, natural pullulan/dextran scaffold that was supplemented with hydroxyapatite crystals that allowed for the spontaneous formation of spheroids. After 2 weeks, their viability was higher under the dynamic conditions (>94%) than the static conditions (<75%), with dead cells central in the spheroids. Mineralization and collagen IV production increased under the dynamic conditions, correlating with osteogenesis and vasculogenesis. The endothelial cells clustered at the spheroidal core by day 7. Proliferation doubled in the dynamic conditions, especially at the scaffold peripheries. Lattice Boltzmann simulations showed negligible wall shear stress in the hydrogel pores but highlighted highly oxygenated zones coinciding with cell proliferation. A strong oxygen gradient likely influenced endothelial migration and cell distribution. Hypoxia was minimal, explaining high viability and spheroid maturation in the dynamic conditions.
Collapse
Affiliation(s)
- Soukaina El Hajj
- Laboratoire de Mécanique Paris-Saclay, CNRS, CentraleSupélec, ENS Paris-Saclay, Université Paris-Saclay, 91190 Gif-sur-Yvette, France;
| | - Martial Bankoué Ntaté
- Laboratoire de Génie des Procédés et Matériaux, CentraleSupélec, Université Paris-Saclay, 91190 Gif-sur-Yvette, France; (M.B.N.); (C.B.); (M.D.); (H.D.)
| | - Cyril Breton
- Laboratoire de Génie des Procédés et Matériaux, CentraleSupélec, Université Paris-Saclay, 91190 Gif-sur-Yvette, France; (M.B.N.); (C.B.); (M.D.); (H.D.)
| | - Robin Siadous
- Laboratoire de Bioingénierie Tissulaire (BioTis), INSERM U1026, Université de Bordeaux, 33076 Bordeaux, France; (R.S.); (J.A.)
| | - Rachida Aid
- Laboratoire de Recherche Vasculaire Translationnelle (LVTS), INSERM U1148, Université Paris Cité, 75018 Paris, France; (R.A.); (D.L.)
- Laboratoire de Recherche Vasculaire Translationnelle (LVTS), INSERM U1148, Université Sorbonne Paris Nord, 93430 Villetaneuse, France
| | - Magali Dupuy
- Laboratoire de Génie des Procédés et Matériaux, CentraleSupélec, Université Paris-Saclay, 91190 Gif-sur-Yvette, France; (M.B.N.); (C.B.); (M.D.); (H.D.)
| | - Didier Letourneur
- Laboratoire de Recherche Vasculaire Translationnelle (LVTS), INSERM U1148, Université Paris Cité, 75018 Paris, France; (R.A.); (D.L.)
- Laboratoire de Recherche Vasculaire Translationnelle (LVTS), INSERM U1148, Université Sorbonne Paris Nord, 93430 Villetaneuse, France
| | - Joëlle Amédée
- Laboratoire de Bioingénierie Tissulaire (BioTis), INSERM U1026, Université de Bordeaux, 33076 Bordeaux, France; (R.S.); (J.A.)
| | - Hervé Duval
- Laboratoire de Génie des Procédés et Matériaux, CentraleSupélec, Université Paris-Saclay, 91190 Gif-sur-Yvette, France; (M.B.N.); (C.B.); (M.D.); (H.D.)
| | - Bertrand David
- Laboratoire de Mécanique Paris-Saclay, CNRS, CentraleSupélec, ENS Paris-Saclay, Université Paris-Saclay, 91190 Gif-sur-Yvette, France;
| |
Collapse
|
2
|
Sun Y, Chen P, Zhao B. Role of extracellular vesicles associated with microRNAs and their interplay with cuproptosis in osteoporosis. Noncoding RNA Res 2024; 9:715-719. [PMID: 38577024 PMCID: PMC10990744 DOI: 10.1016/j.ncrna.2024.03.002] [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: 09/17/2023] [Revised: 03/02/2024] [Accepted: 03/05/2024] [Indexed: 04/06/2024] Open
Abstract
Osteoporosis (OP)-associated fractures can result in severe morbidity and disability, reduced quality of life, and death. Previous studies have suggested that small noncoding RNAs, for example, small regulatory microRNAs (miRNAs), play a key role in OP by inhibiting target gene expression. Cuproptosis, a recently proposed copper-induced cell death pathway, is linked with OP. Here, we describe the contribution of exosomal miRNAs and cuproptosis to OP. First, we highlight the characteristics of exosomes and roles of exosome-related miRNAs. Next, we discuss the relationship between cuproptosis and OP. Subsequently, we analyze the crosstalk of exosomal miRNAs with cuproptosis in the development of OP. This review aims to investigate a new clinical treatment method for OP.
Collapse
Affiliation(s)
- Yong Sun
- Department of Sports Medicine, Fourth Medical Center of Chinese PLA General Hospital, Beijing, 100853, China
| | - Peng Chen
- Department of Orthopedics, The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong Province, China
| | - Bin Zhao
- Department of Sports Medicine, Fourth Medical Center of Chinese PLA General Hospital, Beijing, 100853, China
| |
Collapse
|
3
|
Almeida-Pinto J, Moura BS, Gaspar VM, Mano JF. Advances in Cell-Rich Inks for Biofabricating Living Architectures. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2313776. [PMID: 38639337 DOI: 10.1002/adma.202313776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Revised: 04/15/2024] [Indexed: 04/20/2024]
Abstract
Advancing biofabrication toward manufacturing living constructs with well-defined architectures and increasingly biologically relevant cell densities is highly desired to mimic the biofunctionality of native human tissues. The formulation of tissue-like, cell-dense inks for biofabrication remains, however, challenging at various levels of the bioprinting process. Promising advances have been made toward this goal, achieving relatively high cell densities that surpass those found in conventional platforms, pushing the current boundaries closer to achieving tissue-like cell densities. On this focus, herein the overarching challenges in the bioprocessing of cell-rich living inks into clinically grade engineered tissues are discussed, as well as the most recent advances in cell-rich living ink formulations and their processing technologies are highlighted. Additionally, an overview of the foreseen developments in the field is provided and critically discussed.
Collapse
Affiliation(s)
- José Almeida-Pinto
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, Campus Universitário de Santiago, Aveiro, 3810-193, Portugal
| | - Beatriz S Moura
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, Campus Universitário de Santiago, Aveiro, 3810-193, Portugal
| | - Vítor M Gaspar
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, Campus Universitário de Santiago, Aveiro, 3810-193, Portugal
| | - João F Mano
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, Campus Universitário de Santiago, Aveiro, 3810-193, Portugal
| |
Collapse
|
4
|
Chen Z, Cao Y, Jiang W, Yan Z, Cai G, Ye J, Wang H, Liu L. Porphyromonas gingivalis OMVs promoting endothelial dysfunction via the STING pathway in periodontitis. Oral Dis 2024. [PMID: 38696515 DOI: 10.1111/odi.14969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 04/06/2024] [Accepted: 04/09/2024] [Indexed: 05/04/2024]
Abstract
OBJECTIVE This study aimed to assess the effects of Porphyromonas gingivalis outer membrane vesicles (Pg-OMVs) in chronic periodontitis and explore the underlying mechanism involved. METHODS In vitro, Pg-OMVs were incubated with Ea.hy926 (vessel endothelial cells, ECs) to evaluate their effects on endothelial functions and to investigate the underlying mechanism. The effects of endothelial dysfunction on MG63 osteoblast-like cells were verified using an indirect co-culture method. For in vivo studies, micro-CT was conducted to identify alveolar bone mass. Immunofluorescence staining was conducted to confirm the levels of stimulator of interferon genes (STING) in the blood vessel and the number of Runx2+ cells around the alveolar bone. RESULTS Pg-OMVs were endocytosed by ECs, leading to endothelial dysfunction. The cGAS-STING-TBK1 pathway was activated in ECs, which subsequently inhibited MG63 migration and early osteogenesis differentiation. In vivo, Pg-OMVs promoted alveolar bone resorption, increased STING levels in the blood vessel, and decreased Runx2+ cells around the alveolar bone. CONCLUSIONS Pg-OMVs caused endothelial dysfunction and activated the cGAS-STING-TBK1 signal cascade in ECs, thereby impairing ECs-mediated osteogenesis. Furthermore, Pg-OMVs aggregated alveolar bone loss and altered the blood vessel-mediated osteogenesis with elevated STING.
Collapse
Affiliation(s)
- Zhenwei Chen
- Department of Orthodontics, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China
- State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases (Nanjing Medical University), Nanjing, China
| | - Ye Cao
- Department of Orthodontics, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China
- State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases (Nanjing Medical University), Nanjing, China
| | - Wenxiu Jiang
- Department of Orthodontics, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China
- State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases (Nanjing Medical University), Nanjing, China
| | - Zixin Yan
- Department of Orthodontics, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China
- State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases (Nanjing Medical University), Nanjing, China
| | - Guanhui Cai
- Department of Orthodontics, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China
- State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases (Nanjing Medical University), Nanjing, China
| | - Junjie Ye
- Department of Orthodontics, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China
- State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases (Nanjing Medical University), Nanjing, China
| | - Hua Wang
- Department of Orthodontics, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China
- State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases (Nanjing Medical University), Nanjing, China
- Department of Orthodontics, Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing, China
| | - Luwei Liu
- Department of Orthodontics, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China
- State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases (Nanjing Medical University), Nanjing, China
- Department of Orthodontics, Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing, China
| |
Collapse
|
5
|
Yang J, Chen Z, Gao C, Liu J, Liu K, Wang X, Pan X, Wang G, Sang H, Pan H, Liu W, Ruan C. A mechanical-assisted post-bioprinting strategy for challenging bone defects repair. Nat Commun 2024; 15:3565. [PMID: 38670999 PMCID: PMC11053166 DOI: 10.1038/s41467-024-48023-8] [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: 07/26/2023] [Accepted: 04/17/2024] [Indexed: 04/28/2024] Open
Abstract
Bioprinting that can synchronously deposit cells and biomaterials has lent fresh impetus to the field of tissue regeneration. However, the unavoidable occurrence of cell damage during fabrication process and intrinsically poor mechanical stability of bioprinted cell-laden scaffolds severely restrict their utilization. As such, on basis of heart-inspired hollow hydrogel-based scaffolds (HHSs), a mechanical-assisted post-bioprinting strategy is proposed to load cells into HHSs in a rapid, uniform, precise and friendly manner. HHSs show mechanical responsiveness to load cells within 4 s, a 13-fold increase in cell number, and partitioned loading of two types of cells compared with those under static conditions. As a proof of concept, HHSs with the loading cells show an enhanced regenerative capability in repair of the critical-sized segmental and osteoporotic bone defects in vivo. We expect that this post-bioprinting strategy can provide a universal, efficient, and promising way to promote cell-based regenerative therapy.
Collapse
Affiliation(s)
- Jirong Yang
- Research Center for Human Tissue and Organ Degeneration, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Zhigang Chen
- Research Center for Human Tissue and Organ Degeneration, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Chongjian Gao
- Research Center for Human Tissue and Organ Degeneration, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Juan Liu
- Research Center for Human Tissue and Organ Degeneration, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Kaizheng Liu
- Research Center for Human Tissue and Organ Degeneration, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Xiao Wang
- Research Center for Human Tissue and Organ Degeneration, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
- Shenzhen Hospital, Southern Medical University, Shenzhen, 518000, China
| | - Xiaoling Pan
- Research Center for Human Tissue and Organ Degeneration, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
- Shenzhen Hospital, Southern Medical University, Shenzhen, 518000, China
| | - Guocheng Wang
- Research Center for Human Tissue and Organ Degeneration, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Hongxun Sang
- Shenzhen Hospital, Southern Medical University, Shenzhen, 518000, China
| | - Haobo Pan
- Research Center for Human Tissue and Organ Degeneration, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
- The Key Laboratory of Biomedical Imaging Science and System, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Wenguang Liu
- School of Materials Science and Engineering, Tianjin Key Laboratory of Composite and Functional Materials, Tianjin University, Tianjin, 300350, China
| | - Changshun Ruan
- Research Center for Human Tissue and Organ Degeneration, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- The Key Laboratory of Biomedical Imaging Science and System, Chinese Academy of Sciences, Shenzhen, 518055, China.
| |
Collapse
|
6
|
Chen W, Wang Q, Tao H, Lu L, Zhou J, Wang Q, Huang W, Yang X. Subchondral osteoclasts and osteoarthritis: new insights and potential therapeutic avenues. Acta Biochim Biophys Sin (Shanghai) 2024; 56:499-512. [PMID: 38439665 DOI: 10.3724/abbs.2024017] [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] [Indexed: 03/06/2024] Open
Abstract
Osteoarthritis (OA) is the most common joint disease, and good therapeutic results are often difficult to obtain due to its complex pathogenesis and diverse causative factors. After decades of research and exploration of OA, it has been progressively found that subchondral bone is essential for its pathogenesis, and pathological changes in subchondral bone can be observed even before cartilage lesions develop. Osteoclasts, the main cells regulating bone resorption, play a crucial role in the pathogenesis of subchondral bone. Subchondral osteoclasts regulate the homeostasis of subchondral bone through the secretion of degradative enzymes, immunomodulation, and cell signaling pathways. In OA, osteoclasts are overactivated by autophagy, ncRNAs, and Rankl/Rank/OPG signaling pathways. Excessive bone resorption disrupts the balance of bone remodeling, leading to increased subchondral bone loss, decreased bone mineral density and consequent structural damage to articular cartilage and joint pain. With increased understanding of bone biology and targeted therapies, researchers have found that the activity and function of subchondral osteoclasts are affected by multiple pathways. In this review, we summarize the roles and mechanisms of subchondral osteoclasts in OA, enumerate the latest advances in subchondral osteoclast-targeted therapy for OA, and look forward to the future trends of subchondral osteoclast-targeted therapies in clinical applications to fill the gaps in the current knowledge of OA treatment and to develop new therapeutic strategies.
Collapse
Affiliation(s)
- Wenlong Chen
- Orthopedics and Sports Medicine Center, Suzhou Municipal Hospital, Nanjing Medical University Affiliated Suzhou Hospital, Suzhou 215000, China
- Gusu School, Nanjing Medical University, Suzhou 215000, China
| | - Qiufei Wang
- Department of Orthopedics, the First Affiliated Hospital of Soochow University, Suzhou 215000, China
| | - Huaqiang Tao
- Department of Orthopedics, the First Affiliated Hospital of Soochow University, Suzhou 215000, China
| | - Lingfeng Lu
- Orthopedics and Sports Medicine Center, Suzhou Municipal Hospital, Nanjing Medical University Affiliated Suzhou Hospital, Suzhou 215000, China
- Gusu School, Nanjing Medical University, Suzhou 215000, China
| | - Jing Zhou
- Orthopedics and Sports Medicine Center, Suzhou Municipal Hospital, Nanjing Medical University Affiliated Suzhou Hospital, Suzhou 215000, China
- Gusu School, Nanjing Medical University, Suzhou 215000, China
| | - Qiang Wang
- Department of Orthopedics, the First Affiliated Hospital of Soochow University, Suzhou 215000, China
| | - Wei Huang
- Department of Orthopaedics, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
| | - Xing Yang
- Orthopedics and Sports Medicine Center, Suzhou Municipal Hospital, Nanjing Medical University Affiliated Suzhou Hospital, Suzhou 215000, China
- Gusu School, Nanjing Medical University, Suzhou 215000, China
| |
Collapse
|
7
|
Luo Y, Liu H, Chen M, Zhang Y, Zheng W, Wu L, Liu Y, Liu S, Luo E, Liu X. Immunomodulatory nanomedicine for osteoporosis: Current practices and emerging prospects. Acta Biomater 2024; 179:13-35. [PMID: 38494082 DOI: 10.1016/j.actbio.2024.03.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 02/22/2024] [Accepted: 03/11/2024] [Indexed: 03/19/2024]
Abstract
Osteoporosis results from the disruption of the balance between bone resorption and bone formation. However, classical anti-osteoporosis drugs exhibit several limitations in clinical applications, such as multiple adverse reactions and poor therapeutic effects. Therefore, there is an urgent need for alternative treatment strategies. With the evolution of immunomodulatory nanomedicine, a variety of nanomaterials have been designed for anti-osteoporosis treatment, offering prospects of minimal adverse reactions, enhanced bone induction, and high osteogenic activity. This review initially provides a brief overview of the fundamental principles of bone reconstruction, current osteogenic clinical methods in osteoporosis treatment, and the significance of osteogenic-angiogenic coupling, laying the groundwork for understanding the pathophysiology and therapeutics of osteoporosis. Subsequently, the article emphasizes the relationship between bone immunity and osteogenesis-angiogenesis coupling and provides a detailed analysis of the application of immunomodulatory nanomedicines in the treatment of osteoporosis, including various types of nanomaterials and their integration with carrier biomaterials. Importantly, we discuss the potential of some emerging strategies in immunomodulatory nanomedicine for osteoporosis treatment. This review introduces the innovative applications of immunomodulatory nanomedicine in the treatment of osteoporosis, aiming to serve as a reference for the application of immunomodulatory nanomedicine strategies in osteoporosis treatment. STATEMENT OF SIGNIFICANCE: Osteoporosis, as one of the most prevalent skeletal disorders, poses a significant threat to public health. To date, conventional anti-osteoporosis strategies have been limited in efficacy and plagued with numerous side effects. Fortunately, with the advancement of research in osteoimmunology and nanomedicine, strategies integrating these two fields show great promise in combating osteoporosis. Nanomedicine with immunomodulatory properties exhibits enhanced efficiency, prolonged effectiveness, and increased safety. However, as of now, there exists no comprehensive review amalgamating immunomodulation with nanomedicine to delineate the progress of immunomodulatory nanomedicine in osteoporosis treatment, as well as the future direction of this strategy.
Collapse
Affiliation(s)
- Yankun Luo
- State Key Laboratory of Oral Diseases & National Center for Stomatology& National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Hanghang Liu
- State Key Laboratory of Oral Diseases & National Center for Stomatology& National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Ming Chen
- West China School of Medicine, Sichuan University, Chengdu 610041, Sichuan, China
| | - Yaowen Zhang
- State Key Laboratory of Oral Diseases & National Center for Stomatology& National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Wenzhuo Zheng
- State Key Laboratory of Oral Diseases & National Center for Stomatology& National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Li Wu
- College of Electronics Information and Engineering, Sichuan University, Chengdu 610064, Sichuan, China
| | - Yao Liu
- State Key Laboratory of Oral Diseases & National Center for Stomatology& National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Shibo Liu
- State Key Laboratory of Oral Diseases & National Center for Stomatology& National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - En Luo
- State Key Laboratory of Oral Diseases & National Center for Stomatology& National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Xian Liu
- State Key Laboratory of Oral Diseases & National Center for Stomatology& National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China.
| |
Collapse
|
8
|
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
|
9
|
Liu H, Chen H, Han Q, Sun B, Liu Y, Zhang A, Fan D, Xia P, Wang J. Recent advancement in vascularized tissue-engineered bone based on materials design and modification. Mater Today Bio 2023; 23:100858. [PMID: 38024843 PMCID: PMC10679779 DOI: 10.1016/j.mtbio.2023.100858] [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: 06/02/2023] [Revised: 09/03/2023] [Accepted: 11/06/2023] [Indexed: 12/01/2023] Open
Abstract
Bone is one of the most vascular network-rich tissues in the body and the vascular system is essential for the development, homeostasis, and regeneration of bone. When segmental irreversible damage occurs to the bone, restoring its vascular system by means other than autogenous bone grafts with vascular pedicles is a therapeutic challenge. By pre-generating the vascular network of the scaffold in vivo or in vitro, the pre-vascularization technique enables an abundant blood supply in the scaffold after implantation. However, pre-vascularization techniques are time-consuming, and in vivo pre-vascularization techniques can be damaging to the body. Critical bone deficiencies may be filled quickly with immediate implantation of a supporting bone tissue engineered scaffold. However, bone tissue engineered scaffolds generally lack vascularization, which requires modification of the scaffold to aid in enhancing internal vascularization. In this review, we summarize the relationship between the vascular system and osteogenesis and use it as a basis to further discuss surgical and cytotechnology-based pre-vascularization strategies and to describe the preparation of vascularized bone tissue engineered scaffolds that can be implanted immediately. We anticipate that this study will serve as inspiration for future vascularized bone tissue engineered scaffold construction and will aid in the achievement of clinical vascularized bone.
Collapse
Affiliation(s)
- Hao Liu
- Department of Orthopedic Surgery, The Second Hospital of Jilin University, Changchun 130000, Jilin, China
| | - Hao Chen
- Department of Orthopedic Surgery, The Second Hospital of Jilin University, Changchun 130000, Jilin, China
| | - Qin Han
- Department of Orthopedic Surgery, The Second Hospital of Jilin University, Changchun 130000, Jilin, China
| | - Bin Sun
- Department of Orthopedic Surgery, The Second Hospital of Jilin University, Changchun 130000, Jilin, China
| | - Yang Liu
- Department of Orthopedic Surgery, The Second Hospital of Jilin University, Changchun 130000, Jilin, China
| | - Aobo Zhang
- Department of Orthopedic Surgery, The Second Hospital of Jilin University, Changchun 130000, Jilin, China
| | - Danyang Fan
- Department of Dermatology, The Second Hospital of Jilin University, Changchun 130000, Jilin, China
| | - Peng Xia
- Department of Orthopedic Surgery, The Second Hospital of Jilin University, Changchun 130000, Jilin, China
| | - Jincheng Wang
- Department of Orthopedic Surgery, The Second Hospital of Jilin University, Changchun 130000, Jilin, China
| |
Collapse
|
10
|
Wang C, Min S, Tian Y. Injectable and Cell-Laden Hydrogel in the Contained Bone Defect Animal Model: A Systematic Review. Tissue Eng Regen Med 2023; 20:829-837. [PMID: 37563482 PMCID: PMC10519912 DOI: 10.1007/s13770-023-00569-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 06/22/2023] [Accepted: 07/03/2023] [Indexed: 08/12/2023] Open
Abstract
BACKGROUND Due to its high water content and biomimetic properties simulating extracellular matrix (ECM), hydrogels have been used as preferred cell culture and delivery systems. Similarly, cell-loaded hydrogels can be easily injected into target areas in a minimally invasive manner, minimizing surgical trauma, adapting to irregular shaped defects, and benefiting patients. In this study, we systematically reviewed multiple studies on hydrogel-based bone defect research and briefly summarized the progress of injectable and cell-loaded hydrogels in bone defect repair. METHODS A systematic search was conducted in the PubMed and Web of Science databases using selected search terms. RESULTS Initially, 185 articles were retrieved from the databases. After full-text screening based on inclusion and exclusion criteria, 26 articles were included in this systematic review. Data collected from each study included culture model, seed cell type and origin, cell concentration, scaffold material, scaffold shape, experimental animal and site, bioactive agents, and binding method. This injectable and cell-loaded hydrogel shows certain feasibility in bone tissue engineering applications. CONCLUSION Injectable and cell-loaded hydrogels have been widely applied in bone tissue engineering research. The future direction of bone tissue engineering for bone defect treatment involves the use of new hydrogel materials and biochemical stimulation.
Collapse
Affiliation(s)
- Chaoxin Wang
- Department of Orthopedics, Peking University Third Hospital, Beijing, 100191, China
- Engineering Research Center of Bone and Joint Precision Medicine, Ministry of Education, Beijing, 100191, China
| | - Shuyuan Min
- Department of Orthopedics, Peking University Third Hospital, Beijing, 100191, China
- Engineering Research Center of Bone and Joint Precision Medicine, Ministry of Education, Beijing, 100191, China
| | - Yun Tian
- Department of Orthopedics, Peking University Third Hospital, Beijing, 100191, China.
- Engineering Research Center of Bone and Joint Precision Medicine, Ministry of Education, Beijing, 100191, China.
| |
Collapse
|
11
|
Wu T, Jiang Y, Shi W, Wang Y, Li T. Endoplasmic reticulum stress: a novel targeted approach to repair bone defects by regulating osteogenesis and angiogenesis. J Transl Med 2023; 21:480. [PMID: 37464413 PMCID: PMC10353205 DOI: 10.1186/s12967-023-04328-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Accepted: 07/06/2023] [Indexed: 07/20/2023] Open
Abstract
Bone regeneration therapy is clinically important, and targeted regulation of endoplasmic reticulum (ER) stress is important in regenerative medicine. The processing of proteins in the ER controls cell fate. The accumulation of misfolded and unfolded proteins occurs in pathological states, triggering ER stress. ER stress restores homeostasis through three main mechanisms, including protein kinase-R-like ER kinase (PERK), inositol-requiring enzyme 1ɑ (IRE1ɑ) and activating transcription factor 6 (ATF6), collectively known as the unfolded protein response (UPR). However, the UPR has both adaptive and apoptotic effects. Modulation of ER stress has therapeutic potential for numerous diseases. Repair of bone defects involves both angiogenesis and bone regeneration. Here, we review the effects of ER stress on osteogenesis and angiogenesis, with emphasis on ER stress under high glucose (HG) and inflammatory conditions, and the use of ER stress inducers or inhibitors to regulate osteogenesis and angiogenesis. In addition, we highlight the ability for exosomes to regulate ER stress. Recent advances in the regulation of ER stress mediated osteogenesis and angiogenesis suggest novel therapeutic options for bone defects.
Collapse
Affiliation(s)
- Tingyu Wu
- Department of Joint Surgery, The Affiliated Hospital of Qingdao University, No. 59, Haier Road, Qingdao, 266003, China
| | - Yaping Jiang
- Department of Oral Implantology, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
| | - Weipeng Shi
- Department of Joint Surgery, The Affiliated Hospital of Qingdao University, No. 59, Haier Road, Qingdao, 266003, China
| | - Yingzhen Wang
- Department of Joint Surgery, The Affiliated Hospital of Qingdao University, No. 59, Haier Road, Qingdao, 266003, China
| | - Tao Li
- Department of Joint Surgery, The Affiliated Hospital of Qingdao University, No. 59, Haier Road, Qingdao, 266003, China.
| |
Collapse
|
12
|
Mahapatra C, Kumar P, Paul MK, Kumar A. Angiogenic stimulation strategies in bone tissue regeneration. Tissue Cell 2022; 79:101908. [DOI: 10.1016/j.tice.2022.101908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 03/24/2022] [Accepted: 08/22/2022] [Indexed: 11/28/2022]
|
13
|
Shan N, Lu Y, Guo H, Li D, Jiang J, Yan L, Gao J, Ren Y, Zhao X, Hou L. CITEdb: a manually curated database of cell-cell interactions in human. Bioinformatics 2022; 38:5144-5148. [PMID: 36179089 PMCID: PMC9665858 DOI: 10.1093/bioinformatics/btac654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 08/31/2022] [Accepted: 09/29/2022] [Indexed: 12/24/2022] Open
Abstract
MOTIVATION The interactions among various types of cells play critical roles in cell functions and the maintenance of the entire organism. While cell-cell interactions are traditionally revealed from experimental studies, recent developments in single-cell technologies combined with data mining methods have enabled computational prediction of cell-cell interactions, which have broadened our understanding of how cells work together, and have important implications in therapeutic interventions targeting cell-cell interactions for cancers and other diseases. Despite the importance, to our knowledge, there is no database for systematic documentation of high-quality cell-cell interactions at the cell type level, which hinders the development of computational approaches to identify cell-cell interactions. RESULTS We develop a publicly accessible database, CITEdb (Cell-cell InTEraction database, https://citedb.cn/), which not only facilitates interactive exploration of cell-cell interactions in specific physiological contexts (e.g. a disease or an organ) but also provides a benchmark dataset to interpret and evaluate computationally derived cell-cell interactions from different tools. CITEdb contains 728 pairs of cell-cell interactions in human that are manually curated. Each interaction is equipped with structured annotations including the physiological context, the ligand-receptor pairs that mediate the interaction, etc. Our database provides a web interface to search, visualize and download cell-cell interactions. Users can search for cell-cell interactions by selecting the physiological context of interest or specific cell types involved. CITEdb is the first attempt to catalogue cell-cell interactions at the cell type level, which is beneficial to both experimental, computational and clinical studies of cell-cell interactions. AVAILABILITY AND IMPLEMENTATION CITEdb is freely available at https://citedb.cn/ and the R package implementing benchmark is available at https://github.com/shanny01/benchmark. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Nayang Shan
- School of Statistics, Capital University of Economics and Business, Beijing 100070, China
| | - Yao Lu
- Department of Industrial Engineering, Center for Statistical Science, Tsinghua University, Beijing 100084, China
- MOE Key Laboratory of Bioinformatics, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Hao Guo
- State Key Laboratory of Translational Medicine and Innovative Drug Development, Jiangsu Simcere Diagnostics Co., Ltd, Nanjing 210042, China
- Nanjing Simcere Medical Laboratory Science Co., Ltd, Nanjing 210042, China
| | - Dongyu Li
- Department of Industrial Engineering, Center for Statistical Science, Tsinghua University, Beijing 100084, China
- MOE Key Laboratory of Bioinformatics, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Jitong Jiang
- Department of Mathematics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Linlin Yan
- State Key Laboratory of Translational Medicine and Innovative Drug Development, Jiangsu Simcere Diagnostics Co., Ltd, Nanjing 210042, China
- Nanjing Simcere Medical Laboratory Science Co., Ltd, Nanjing 210042, China
| | - Jiudong Gao
- State Key Laboratory of Translational Medicine and Innovative Drug Development, Jiangsu Simcere Diagnostics Co., Ltd, Nanjing 210042, China
- Nanjing Simcere Medical Laboratory Science Co., Ltd, Nanjing 210042, China
| | - Yong Ren
- State Key Laboratory of Translational Medicine and Innovative Drug Development, Jiangsu Simcere Diagnostics Co., Ltd, Nanjing 210042, China
- Nanjing Simcere Medical Laboratory Science Co., Ltd, Nanjing 210042, China
| | - Xingming Zhao
- Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence (LCNBI) and ZJLab, Wuxi, China
| | - Lin Hou
- Department of Industrial Engineering, Center for Statistical Science, Tsinghua University, Beijing 100084, China
- MOE Key Laboratory of Bioinformatics, School of Life Sciences, Tsinghua University, Beijing 100084, China
| |
Collapse
|
14
|
Che Z, Song Y, Zhu L, Liu T, Li X, Huang L. Emerging roles of growth factors in osteonecrosis of the femoral head. Front Genet 2022; 13:1037190. [PMID: 36452155 PMCID: PMC9702520 DOI: 10.3389/fgene.2022.1037190] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 10/24/2022] [Indexed: 12/20/2023] Open
Abstract
Osteonecrosis of the femoral head (ONFH) is a potentially disabling orthopedic condition that requires total hip arthroplasty in most late-stage cases. However, mechanisms underlying the development of ONFH remain unknown, and the therapeutic strategies remain limited. Growth factors play a crucial role in different physiological processes, including cell proliferation, invasion, metabolism, apoptosis, and stem cell differentiation. Recent studies have reported that polymorphisms of growth factor-related genes are involved in the pathogenesis of ONFH. Tissue and genetic engineering are attractive strategies for treating early-stage ONFH. In this review, we summarized dysregulated growth factor-related genes and their role in the occurrence and development of ONFH. In addition, we discussed their potential clinical applications in tissue and genetic engineering for the treatment of ONFH.
Collapse
Affiliation(s)
- Zhenjia Che
- Department of Orthopaedics, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Yang Song
- Department of Orthopaedics, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Liwei Zhu
- Department of Orthopaedics, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Tengyue Liu
- Department of Orthopaedics, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Xudong Li
- Department of Orthopaedics, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Lanfeng Huang
- Department of Orthopaedics, The Second Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
15
|
Banerjee D, Singh YP, Datta P, Ozbolat V, O'Donnell A, Yeo M, Ozbolat IT. Strategies for 3D bioprinting of spheroids: A comprehensive review. Biomaterials 2022; 291:121881. [DOI: 10.1016/j.biomaterials.2022.121881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 10/04/2022] [Accepted: 10/23/2022] [Indexed: 11/17/2022]
|
16
|
Highly porous multiple-cell-laden collagen/hydroxyapatite scaffolds for bone tissue engineering. Int J Biol Macromol 2022; 222:1264-1276. [DOI: 10.1016/j.ijbiomac.2022.09.249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 09/16/2022] [Accepted: 09/27/2022] [Indexed: 11/15/2022]
|
17
|
Tan G, Chen R, Tu X, Guo L, Guo L, Xu J, Zhang C, Zou T, Sun S, Jiang Q. Research on the osteogenesis and biosafety of ECM–Loaded 3D–Printed Gel/SA/58sBG scaffolds. Front Bioeng Biotechnol 2022; 10:973886. [PMID: 36061449 PMCID: PMC9438739 DOI: 10.3389/fbioe.2022.973886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 07/14/2022] [Indexed: 11/13/2022] Open
Abstract
Employing scaffolds containing cell–derived extracellular matrix (ECM) as an alternative strategy for the regeneration of bone defects has shown prominent advantages. Here, gelatin (Gel), sodium alginate (SA) and 58s bioactive glass (58sBG) were incorporated into deionized water to form ink, which was further fabricated into composite scaffolds by the 3D printing technique. Then, rat aortic endothelial cells (RAOECs) or rat bone mesenchymal stem cells (RBMSCs) were seeded on the scaffolds. After decellularization, two kinds of ECM–loaded scaffolds (RAOECs–ECM scaffold and RBMSCs–ECM scaffold) were obtained. The morphological characteristics of the scaffolds were assessed meticulously by scanning electron microscopy (SEM). In addition, the effects of scaffolds on the proliferation, adhesion, and osteogenic and angiogenic differentiation of RBMSCs were evaluated by Calcein AM staining and reverse transcription polymerase chain reaction (RT–PCR). In vivo, full–thickness bone defects with a diameter of 5 mm were made in the mandibles of Sprague–Dawley (SD) rats to assess the bone regeneration ability and biosafety of the scaffolds at 4, 8 and 16 weeks. The osteogenic and angiogenic potential of the scaffolds were investigated by microcomputed tomography (Micro–CT) and histological analysis. The biosafety of the scaffolds was evaluated by blood biochemical indices and histological staining of the liver, kidney and cerebrum. The results showed that the ECM–loaded scaffolds were successfully prepared, exhibiting interconnected pores and a gel–like ECM distributed on their surfaces. Consistently, in vitro experiments demonstrated that the scaffolds displayed favourable cytocompatibility. In vitro osteogenic differentiation studies showed that scaffolds coated with ECM could significantly increase the expression of osteogenic and angiogenic genes. In addition, the results from mandibular defect repair in vivo revealed that the ECM–loaded scaffolds effectively promoted the healing of bone defects when compared to the pure scaffold. Overall, these findings demonstrate that both RAOECs–ECM scaffold and RBMSCs–ECM scaffold can greatly enhance bone formation with good biocompatibility and thus have potential for clinical application in bone regeneration.
Collapse
Affiliation(s)
- Guozhong Tan
- Department of Endodontics, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, China
- Department of Oral and Maxillofacial Surgery, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, China
| | - Rongfeng Chen
- Department of Endodontics, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, China
| | - Xinran Tu
- Department of Endodontics, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, China
| | - Liyang Guo
- Department of Endodontics, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, China
| | - Lvhua Guo
- Department of Endodontics, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, China
| | - Jingyi Xu
- Department of Endodontics, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, China
| | - Chengfei Zhang
- Endodontology, Restorative Dental Sciences, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China
| | - Ting Zou
- Endodontology, Restorative Dental Sciences, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China
| | - Shuyu Sun
- Department of Endodontics, Stomatological Hospital, Southern Medical University, Guangzhou, China
- *Correspondence: Shuyu Sun, ; Qianzhou Jiang,
| | - Qianzhou Jiang
- Department of Endodontics, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, China
- *Correspondence: Shuyu Sun, ; Qianzhou Jiang,
| |
Collapse
|
18
|
Liang X, Xie L, Zhang Q, Wang G, Zhang S, Jiang M, Zhang R, Yang T, Hu X, Yang Z, Tian W. Gelatin methacryloyl-alginate core-shell microcapsules as efficient delivery platforms for prevascularized microtissues in endodontic regeneration. Acta Biomater 2022; 144:242-257. [PMID: 35364321 DOI: 10.1016/j.actbio.2022.03.045] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Revised: 03/17/2022] [Accepted: 03/24/2022] [Indexed: 02/06/2023]
Abstract
Combined injectable cell-laden microspheres and angiogenesis approaches are promising for functional vascularized endodontic regeneration. However, advanced microsphere designs and production techniques that benefit practical applications are rarely developed. Herein, gelatin methacryloyl (GelMA)-alginate core-shell microcapsules were fabricated to co-encapsulate human dental pulp stem cells (hDPSCs) and human umbilical vein endothelial cells (HUVECs) based on a coaxial electrostatic microdroplet technique. This technique enables high-throughput production, convenient collection, and minimal material waste. The average diameter of core-shell microcapsules was ∼359 µm, and that of GelMA cores was ∼278 µm. There were higher proliferation rates for hDPSCs and HUVECs co-encapsulated in the GelMA cores than for hDPSCs or HUVECs monoculture group. HUVECs assembled to form 3D capillary-like networks in co-culture microcapsules. Moreover, HUVECs promoted the osteo/odontogenic differentiation of hDPSCs in microcapsules. After 14 days of cultivation, prevascularized microtissues formed in microcapsules that contained abundant deposited extracellular matrix (ECM); no microcapsule aggregation occurred. In vivo studies confirmed that better microvessel formation and pulp-like tissue regeneration occurred in the co-culture group than in hDPSCs group. Thus, an effective platform for prevascularization microtissue preparation was proposed and showed great promise in endodontic regeneration and tissue engineering applications. STATEMENT OF SIGNIFICANCE: Cell-laden microspheres combined with the proangiogenesis approach are promising in endodontic regeneration. We proposed GelMA-alginate core-shell microcapsules generated via the coaxial electrostatic microdroplet (CEM) method, which utilizes a double-lumen needle to allow for core-shell structures to form. The microcapsules were used for co-culturing hDPSCs and HUVECs to harvest large amounts of prevascularized microtissues, which further showed improved vascularization and pulp-like tissue regeneration in vivo. This CEM method and the microcapsule system have advantages of high-throughput generation, convenient collection, and avoid aggregation during long-term culturing. We proposed a high-effective platform for mass production of prevascularized microtissues, which exhibit great promise in the clinical transformation of endodontic regeneration and other applications in regenerative medicine.
Collapse
Affiliation(s)
- Xi Liang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Engineering Research Center of Oral Translational Medicine, Ministry of Education & National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China; Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Li Xie
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Engineering Research Center of Oral Translational Medicine, Ministry of Education & National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China.
| | - Qingyuan Zhang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Engineering Research Center of Oral Translational Medicine, Ministry of Education & National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China; Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Ge Wang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Engineering Research Center of Oral Translational Medicine, Ministry of Education & National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China; Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Siyuan Zhang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Engineering Research Center of Oral Translational Medicine, Ministry of Education & National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Mingyan Jiang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Engineering Research Center of Oral Translational Medicine, Ministry of Education & National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China; Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Ruitao Zhang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Engineering Research Center of Oral Translational Medicine, Ministry of Education & National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China; Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Ting Yang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Engineering Research Center of Oral Translational Medicine, Ministry of Education & National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China; Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Xingyu Hu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Engineering Research Center of Oral Translational Medicine, Ministry of Education & National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China; Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Ziyang Yang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Engineering Research Center of Oral Translational Medicine, Ministry of Education & National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China; Department of Stomatology, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
| | - Weidong Tian
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Engineering Research Center of Oral Translational Medicine, Ministry of Education & National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China; Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
19
|
Zha K, Tian Y, Panayi AC, Mi B, Liu G. Recent Advances in Enhancement Strategies for Osteogenic Differentiation of Mesenchymal Stem Cells in Bone Tissue Engineering. Front Cell Dev Biol 2022; 10:824812. [PMID: 35281084 PMCID: PMC8904963 DOI: 10.3389/fcell.2022.824812] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 02/08/2022] [Indexed: 12/12/2022] Open
Abstract
Although bone is an organ that displays potential for self-healing after damage, bone regeneration does not occur properly in some cases, and it is still a challenge to treat large bone defects. The development of bone tissue engineering provides a new approach to the treatment of bone defects. Among various cell types, mesenchymal stem cells (MSCs) represent one of the most promising seed cells in bone tissue engineering due to their functions of osteogenic differentiation, immunomodulation, and secretion of cytokines. Regulation of osteogenic differentiation of MSCs has become an area of extensive research over the past few years. This review provides an overview of recent research progress on enhancement strategies for MSC osteogenesis, including improvement in methods of cell origin selection, culture conditions, biophysical stimulation, crosstalk with macrophages and endothelial cells, and scaffolds. This is favorable for further understanding MSC osteogenesis and the development of MSC-based bone tissue engineering.
Collapse
Affiliation(s)
- Kangkang Zha
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, China
| | - Yue Tian
- Department of Military Patient Management, The Second Medical Center & National Clinical Research Center for Geriatric Diseases, Institute of Orthopaedics, Chinese PLA General Hospital, Beijing, China
| | - Adriana C. Panayi
- Division of Plastic Surgery, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, United States
| | - Bobin Mi
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, China
- *Correspondence: Bobin Mi, ; Guohui Liu,
| | - Guohui Liu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, China
- *Correspondence: Bobin Mi, ; Guohui Liu,
| |
Collapse
|
20
|
Petrillo S, Genova T, Chinigò G, Roato I, Scarpellino G, Kopecka J, Altruda F, Tolosano E, Riganti C, Mussano F, Munaron L. Endothelial Cells Promote Osteogenesis by Establishing a Functional and Metabolic Coupling With Human Mesenchymal Stem Cells. Front Physiol 2022; 12:813547. [PMID: 35087424 PMCID: PMC8787057 DOI: 10.3389/fphys.2021.813547] [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: 11/11/2021] [Accepted: 12/14/2021] [Indexed: 11/13/2022] Open
Abstract
Bone formation involves a complex crosstalk between endothelial cells (EC) and osteodifferentiating stem cells. This functional interplay is greatly mediated by the paracrine and autocrine action of soluble factors released at the vasculature-bone interface. This study elucidates the molecular and functional responses triggered by this intimate interaction. In this study, we showed that human dermal microvascular endothelial cells (HMEC) induced the expression of pro-angiogenic factors in stem cells from human exfoliated deciduous teeth (SHED) and sustain their osteo-differentiation at the same time. In contrast, osteodifferentiating SHED increased EC recruitment and promoted the formation of complex vascular networks. Moreover, HMEC enhanced anaerobic glycolysis in proliferating SHED without compromising their ability to undergo the oxidative metabolic shift required for adequate osteo-differentiation. Taken together, these findings provide novel insights into the molecular mechanism underlying the synergistic cooperation between EC and stem cells during bone tissue renewal.
Collapse
Affiliation(s)
- Sara Petrillo
- Molecular Biotechnology Center (MBC), Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Tullio Genova
- Department of Life Sciences and Systems Biology, University of Turin, Turin, Italy
| | - Giorgia Chinigò
- Department of Life Sciences and Systems Biology, University of Turin, Turin, Italy
| | - Ilaria Roato
- Department of Surgical Sciences, C.I.R. Dental School, University of Turin, Turin, Italy
| | - Giorgia Scarpellino
- Department of Life Sciences and Systems Biology, University of Turin, Turin, Italy
| | - Joanna Kopecka
- Department of Oncology, University of Turin, Turin, Italy
| | - Fiorella Altruda
- Molecular Biotechnology Center (MBC), Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Emanuela Tolosano
- Molecular Biotechnology Center (MBC), Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Chiara Riganti
- Department of Oncology, University of Turin, Turin, Italy
| | - Federico Mussano
- Department of Surgical Sciences, C.I.R. Dental School, University of Turin, Turin, Italy
| | - Luca Munaron
- Department of Life Sciences and Systems Biology, University of Turin, Turin, Italy
| |
Collapse
|
21
|
Shahabipour F, Tavafoghi M, Aninwene GE, Bonakdar S, Oskuee RK, Shokrgozar MA, Potyondy T, Alambeigi F, Ahadian S. Coaxial 3D bioprinting of tri-polymer scaffolds to improve the osteogenic and vasculogenic potential of cells in co-culture models. J Biomed Mater Res A 2022; 110:1077-1089. [PMID: 35025130 DOI: 10.1002/jbm.a.37354] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Revised: 12/20/2021] [Accepted: 12/27/2021] [Indexed: 12/20/2022]
Abstract
The crosstalk between osteoblasts and endothelial cells is critical for bone vascularization and regeneration. Here, we used a coaxial 3D bioprinting method to directly print an osteon-like structure by depositing angiogenic and osteogenic bioinks from the core and shell regions of the coaxial nozzle, respectively. The bioinks were made up of gelatin, gelatin methacryloyl (GelMA), alginate, and hydroxyapatite (HAp) nanoparticles and were loaded with human umbilical vascular endothelial cells (HUVECs) and osteoblasts (MC3T3) in the core and shell regions, respectively. Conventional monoaxial 3D bioprinting was used as a control method, where the hydrogels, HAp nanoparticles, MC3T3 cells, and HUVECs were all mixed in one bioink and printed from the core nozzle. As a result, the bioprinted scaffolds were composed of cell-laden fibers with either a core-shell or homogenous structure, providing a non-contact (indirect) or contact (direct) co-culture of MC3T3 cells and HUVECs, respectively. Both structures supported the 3D culture of HUVECs and osteoblasts over a long period. The scaffolds also supported the expression of osteogenic and angiogenic factors. However, the gene expression was significantly higher for the core-shell structure than the homogeneous structure due to the well-defined distribution of osteoblasts and endothelial cells and the formation of vessel-like structures in the co-culture system. Our results indicated that the coaxial bioprinting technique, with the ability to create a non-contact co-culture of cells, can provide a more efficient bioprinting strategy for printing highly vascularized and bioactive bone structures.
Collapse
Affiliation(s)
- Fahimeh Shahabipour
- Skin Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,National Cell Bank of Iran, Pasteur Institute of Iran, Tehran, Iran.,Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Maryam Tavafoghi
- Department of Bioengineering, Henry Samueli School of Engineering and Applied Sciences, 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
| | - George E Aninwene
- Department of Bioengineering, Henry Samueli School of Engineering and Applied Sciences, 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.,California NanoSystems Institute (CNSI), University of California-Los Angeles, Los Angeles, California, USA
| | - Shahin Bonakdar
- Skin Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Reza Kazemi Oskuee
- Biomedical Applied Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Tyler Potyondy
- Department of Bioengineering, Henry Samueli School of Engineering and Applied Sciences, 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
| | - Farshid Alambeigi
- Walker Department of Mechanical Engineering, University of Texas at Austin, Austin, Texas, USA
| | - Samad Ahadian
- Terasaki Institute for Biomedical Innovation, Los Angeles, California, USA
| |
Collapse
|
22
|
Guerrero J, Dasen B, Frismantiene A, Pigeot S, Ismail T, Schaefer DJ, Philippova M, Resink TJ, Martin I, Scherberich A. OUP accepted manuscript. Stem Cells Transl Med 2022; 11:213-229. [PMID: 35259280 PMCID: PMC8929526 DOI: 10.1093/stcltm/szab021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 10/31/2021] [Indexed: 11/24/2022] Open
Abstract
Cells of the stromal vascular fraction (SVF) of human adipose tissue have the capacity to generate osteogenic grafts with intrinsic vasculogenic properties. However, cultured adipose-derived stromal cells (ASCs), even after minimal monolayer expansion, lose osteogenic capacity in vivo. Communication between endothelial and stromal/mesenchymal cell lineages has been suggested to improve bone formation and vascularization by engineered tissues. Here, we investigated the specific role of a subpopulation of SVF cells positive for T-cadherin (T-cad), a putative endothelial marker. We found that maintenance during monolayer expansion of a T-cad-positive cell population, composed of endothelial lineage cells (ECs), is mandatory to preserve the osteogenic capacity of SVF cells in vivo and strongly supports their vasculogenic properties. Depletion of T-cad-positive cells from the SVF totally impaired bone formation in vivo and strongly reduced vascularization by SVF cells in association with decreased VEGF and Adiponectin expression. The osteogenic potential of T-cad-depleted SVF cells was fully rescued by co-culture with ECs from a human umbilical vein (HUVECs), constitutively expressing T-cad. Ectopic expression of T-cad in ASCs stimulated mineralization in vitro but failed to rescue osteogenic potential in vivo, indicating that the endothelial nature of the T-cad-positive cells is the key factor for induction of osteogenesis in engineered grafts based on SVF cells. This study demonstrates that crosstalk between stromal and T-cad expressing endothelial cells within adipose tissue critically regulates osteogenesis, with VEGF and adiponectin as associated molecular mediators.
Collapse
Affiliation(s)
- Julien Guerrero
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Boris Dasen
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Agne Frismantiene
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Sebastien Pigeot
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Tarek Ismail
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
- Department of Plastic, Reconstructive, Aesthetic and Hand Surgery, University Hospital Basel, Basel, Switzerland
| | - Dirk J Schaefer
- Department of Plastic, Reconstructive, Aesthetic and Hand Surgery, University Hospital Basel, Basel, Switzerland
| | - Maria Philippova
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Therese J Resink
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Ivan Martin
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Arnaud Scherberich
- Corresponding author: Arnaud Scherberich, Department of Biomedicine, Hebelstrasse 20, University Hospital Basel, 4031 Basel, Switzerland. Tel: +41 061 328 73 75;
| |
Collapse
|
23
|
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: 24] [Impact Index Per Article: 8.0] [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
|
24
|
In Vivo Efficacy of Neutrophil-Mediated Bone Regeneration Using a Rabbit Calvarial Defect Model. Int J Mol Sci 2021; 22:ijms222313016. [PMID: 34884821 PMCID: PMC8657540 DOI: 10.3390/ijms222313016] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 11/26/2021] [Accepted: 11/28/2021] [Indexed: 01/08/2023] Open
Abstract
Reconstruction of bone due to surgical removal or disease-related bony defects is a clinical challenge. It is known that the immune system exerts positive immunomodulatory effects on tissue repair and regeneration. In this study, we evaluated the in vivo efficacy of autologous neutrophils on bone regeneration using a rabbit calvarial defect model. Methods: Twelve rabbits, each with two surgically created calvarial bone defects (10 mm diameter), were randomly divided into two groups; (i) single application of neutrophils (SA-NP) vs. SA-NP control, and (ii) repetitive application of neutrophils (RA-NP) vs. RA-NP control. The animals were euthanized at 4 and 8 weeks post-operatively and the treatment outcomes were evaluated by micro-computed tomography, histology, and histomorphometric analyses. Results: The micro-CT analysis showed a significantly higher bone volume fraction (bone volume/total volume) in the neutrophil-treated groups, i.e., median interquartile range (IQR) SA-NP (18) and RA-NP (24), compared with the untreated controls, i.e., SA-NP (7) and RA-NP (14) at 4 weeks (p < 0.05). Similarly, new bone area fraction (bone area/total area) was significantly higher in neutrophil-treated groups at 4 weeks (p < 0.05). Both SA-NP and RA-NP had a considerably higher bone volume and bone area at 8 weeks, although the difference was not statistically significant. In addition, immunohistochemical analysis at 8 weeks revealed a higher expression of osteocalcin in both SA-NP and RA-NP groups. Conclusions: The present study provides first hand evidence that autologous neutrophils may have a positive effect on promoting new bone formation. Future studies should be performed with a larger sample size in non-human primate models. If proven feasible, this new promising strategy could bring clinical benefits for bone defects to the field of oral and maxillofacial surgery.
Collapse
|
25
|
He CK, Hsu CH. Microfluidic technology for multiple single-cell capture. BIOMICROFLUIDICS 2021; 15:061501. [PMID: 34777676 PMCID: PMC8577867 DOI: 10.1063/5.0057685] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 10/06/2021] [Indexed: 05/25/2023]
Abstract
Microfluidic devices are widely used in single-cell capture and for pairing single cells or groups of cells for cell-cell interaction analysis; these advances have improved drug screening and cell signal transduction analysis. The complex in vivo environment involves interactions between two cells and among multiple cells of the same or different phenotypes. This study reviewed the core principles and performance of several microfluidic multiple- and single-cell capture methods, namely, the microwell, valve, trap, and droplet methods. The advantages and disadvantages of the methods were compared, and suggestions regarding their application to multiple-cell capture were provided. The results may serve as a reference for research on microfluidic multiple single-cell coculture technology.
Collapse
|
26
|
Dalisson B, Charbonnier B, Aoude A, Gilardino M, Harvey E, Makhoul N, Barralet J. Skeletal regeneration for segmental bone loss: Vascularised grafts, analogues and surrogates. Acta Biomater 2021; 136:37-55. [PMID: 34626818 DOI: 10.1016/j.actbio.2021.09.053] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 09/25/2021] [Accepted: 09/28/2021] [Indexed: 02/08/2023]
Abstract
Massive segmental bone defects (SBD) are mostly treated by removing the fibula and transplanting it complete with blood supply. While revolutionary 50 years ago, this remains the standard treatment. This review considers different strategies to repair SBD and emerging potential replacements for this highly invasive procedure. Prior to the technical breakthrough of microsurgery, researchers in the 1960s and 1970s had begun to make considerable progress in developing non autologous routes to repairing SBD. While the breaktthrough of vascularised bone transplantation solved the immediate problem of a lack of reliable repair strategies, much of their prior work is still relevant today. We challenge the assumption that mimicry is necessary or likely to be successful and instead point to the utility of quite crude (from a materials technology perspective), approaches. Together there are quite compelling indications that the body can regenerate entire bone segments with few or no exogenous factors. This is important, as there is a limit to how expensive a bone repair can be and still be widely available to all patients since cost restraints within healthcare systems are not likely to diminish in the near future. STATEMENT OF SIGNIFICANCE: This review is significant because it is a multidisciplinary view of several surgeons and scientists as to what is driving improvement in segmental bone defect repair, why many approaches to date have not succeeded and why some quite basic approaches can be as effective as they are. While there are many reviews of the literature of grafting and bone repair the relative lack of substantial improvement and slow rate of progress in clinical translation is often overlooked and we seek to challenge the reader to consider the issue more broadly.
Collapse
|
27
|
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]
|
28
|
Kim H, Shin YM, Chung S, Kim D, Park DB, Baek S, Park J, Kim SY, Kim D, Yi SW, Lee S, Lee JB, Ko J, Im G, Kang M, Sung H. Cell-Membrane-Derived Nanoparticles with Notch-1 Suppressor Delivery Promote Hypoxic Cell-Cell Packing and Inhibit Angiogenesis Acting as a Two-Edged Sword. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2021; 33:e2101558. [PMID: 34431568 PMCID: PMC11468545 DOI: 10.1002/adma.202101558] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 07/02/2021] [Indexed: 06/13/2023]
Abstract
Cell-cell interactions regulate intracellular signaling via reciprocal contacts of cell membranes in tissue regeneration and cancer growth, indicating a critical need of membrane-derived tools in studying these processes. Hence, cell-membrane-derived nanoparticles (CMNPs) are produced using tonsil-derived mesenchymal stem cells (TMSCs) from children owing to their short doubling time. As target cell types, laryngeal cancer cells are compared to bone-marrow-derived MSCs (BMSCs) because of their cartilage damaging and chondrogenic characteristics, respectively. Treating spheroids of these cell types with CMNPs exacerbates interspheroid hypoxia with robust maintenance of the cell-cell interaction signature for 7 days. Both cell types prefer a hypoxic environment, as opposed to blood vessel formation that is absent in cartilage but is required for cancer growth. Hence, angiogenesis is inhibited by displaying the Notch-1 aptamer on CMNPs. Consequently, laryngeal cancer growth is suppressed efficiently in contrast to improved chondroprotection observed in a series of cell and animal experiments using a xenograft mouse model of laryngeal cancer. Altogether, CMNPs execute a two-edged sword function of inducing hypoxic cell-cell packing, followed by suppressing angiogenesis to promote laryngeal cancer death and chondrogenesis simultaneously. This study presents a previously unexplored therapeutic strategy for anti-cancer and chondroprotective treatment using CMNPs.
Collapse
Affiliation(s)
- Hye‐Seon Kim
- Department of Medical EngineeringYonsei University College of MedicineSeoul03722Republic of Korea
| | - Young Min Shin
- Department of Medical EngineeringYonsei University College of MedicineSeoul03722Republic of Korea
| | - Seyong Chung
- Department of Medical EngineeringYonsei University College of MedicineSeoul03722Republic of Korea
| | - Dahee Kim
- Department of OtorhinolaryngologyYonsei University College of MedicineSeoul03722Republic of Korea
| | - Dan Bi Park
- TMD LAB Co., Ltd6th Floor, 31, Gwangnaru‐ro 8‐gilSeongdong‐guSeoul04799Republic of Korea
| | - Sewoom Baek
- Department of Medical EngineeringYonsei University College of MedicineSeoul03722Republic of Korea
| | - Jeongeun Park
- Department of Medical EngineeringYonsei University College of MedicineSeoul03722Republic of Korea
| | - Si Yeong Kim
- Department of Medical EngineeringYonsei University College of MedicineSeoul03722Republic of Korea
| | - Dae‐Hyun Kim
- Department of Veterinary SurgeryCollege of Veterinary MedicineChungnam National University99, Daehak‐roYuseong‐guDaejeon34134Republic of Korea
| | - Se Won Yi
- TMD LAB Co., Ltd6th Floor, 31, Gwangnaru‐ro 8‐gilSeongdong‐guSeoul04799Republic of Korea
| | - Songhyun Lee
- Department of Medical EngineeringYonsei University College of MedicineSeoul03722Republic of Korea
| | - Jung Bok Lee
- Department of Biological ScienceSookmyung Women's UniversityCheongpa‐ro 47‐gil 100, Yongsan‐guSeoul04310Republic of Korea
| | - Ji‐Yun Ko
- Department of Veterinary SurgeryCollege of Veterinary MedicineChungnam National University99, Daehak‐roYuseong‐guDaejeon34134Republic of Korea
- Research Institute of Convergence Life ScienceDongguk UniversityGoyang10326Republic of Korea
| | - Gun‐Il Im
- Department of OrthopedicsDongguk University Ilsan HospitalGoyang10326Republic of Korea
| | - Mi‐Lan Kang
- TMD LAB Co., Ltd6th Floor, 31, Gwangnaru‐ro 8‐gilSeongdong‐guSeoul04799Republic of Korea
| | - Hak‐Joon Sung
- Department of Medical EngineeringYonsei University College of MedicineSeoul03722Republic of Korea
| |
Collapse
|
29
|
Rebuilding the hematopoietic stem cell niche: Recent developments and future prospects. Acta Biomater 2021; 132:129-148. [PMID: 33813090 DOI: 10.1016/j.actbio.2021.03.061] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 03/25/2021] [Accepted: 03/25/2021] [Indexed: 12/20/2022]
Abstract
Hematopoietic stem cells (HSCs) have proven their clinical relevance in stem cell transplantation to cure patients with hematological disorders. Key to their regenerative potential is their natural microenvironment - their niche - in the bone marrow (BM). Developments in the field of biomaterials enable the recreation of such environments with increasing preciseness in the laboratory. Such artificial niches help to gain a fundamental understanding of the biophysical and biochemical processes underlying the interaction of HSCs with the materials in their environment and the disturbance of this interplay during diseases affecting the BM. Artificial niches also have the potential to multiply HSCs in vitro, to enable the targeted differentiation of HSCs into mature blood cells or to serve as drug-testing platforms. In this review, we will introduce the importance of artificial niches followed by the biology and biophysics of the natural archetype. We will outline how 2D biomaterials can be used to dissect the complexity of the natural niche into individual parameters for fundamental research and how 3D systems evolved from them. We will present commonly used biomaterials for HSC research and their applications. Finally, we will highlight two areas in the field of HSC research, which just started to unlock the possibilities provided by novel biomaterials, in vitro blood production and studying the pathophysiology of the niche in vitro. With these contents, the review aims to give a broad overview of the different biomaterials applied for HSC research and to discuss their potentials, challenges and future directions in the field. STATEMENT OF SIGNIFICANCE: Hematopoietic stem cells (HSCs) are multipotent cells responsible for maintaining the turnover of all blood cells. They are routinely applied to treat patients with hematological diseases. This high clinical relevance explains the necessity of multiplication or differentiation of HSCs in the laboratory, which is hampered by the missing natural microenvironment - the so called niche. Biomaterials offer the possibility to mimic the niche and thus overcome this hurdle. The review introduces the HSC niche in the bone marrow and discusses the utility of biomaterials in creating artificial niches. It outlines how 2D systems evolved into sophisticated 3D platforms, which opened the gateway to applications such as, expansion of clinically relevant HSCs, in vitro blood production, studying niche pathologies and drug testing.
Collapse
|
30
|
Shi Z, Wang S, Deng J, Gong Z. PGC-1α attenuates the oxidative stress-induced impaired osteogenesis and angiogenesis regulation effects of mesenchymal stem cells in the presence of diabetic serum. Biochem Biophys Rep 2021; 27:101070. [PMID: 34286110 PMCID: PMC8278528 DOI: 10.1016/j.bbrep.2021.101070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 06/28/2021] [Accepted: 06/29/2021] [Indexed: 11/30/2022] Open
Abstract
Oxidative stress is believed to induce dysfunction of the bone remodeling process and be associated with progressive loss of bone mass. The peroxisome proliferator-activated receptor gamma coactivator 1α (PGC-1α) is a master controller during mitochondrial biogenesis and the antioxidant response. We postulated that PGC-1α could function as a cyto-protective effector in mesenchymal stem cells (MSCs) under oxidative stress conditions. In this study, diabetic serum was firstly used to treat MSCs to induce oxidative damage. The anti-oxidative protective effects of PGC-1α overexpression on MSCs, as well as MSCs' osteogenesis and angiogenic regulation effects were investigated in vitro. Results showed that diabetic conditions induced significantly increase of intracellular oxidative damage and mitochondrial permeability transition pore (mPTP) opening activity, decrease of cellular viability, and osteogenic differentiation and pro-angiogenic regulation effects of MSCs. However, the diabetic conditions induced oxidative impair on MSCs were significantly alleviated via PGC-1α overexpression under diabetic conditions. Taken together, this study indicates the anti-oxidative treatment potential of PGC-1α regulation as a promising strategy to promote coupling pro-osteogenesis and pro-angiogenesis effects of MSCs.
Collapse
Affiliation(s)
- Zongxin Shi
- Department of Orthopedic Surgery, Liangxiang Hospital of Beijing Fangshan District, and Liangxiang Teaching Hospital of Capital Medical University, No.45, Gongchen Ave., Liangxiang, Fangshan Dist., Beijing, 102488, China
| | - Shikun Wang
- Department of Orthopedic Surgery, Liangxiang Hospital of Beijing Fangshan District, and Liangxiang Teaching Hospital of Capital Medical University, No.45, Gongchen Ave., Liangxiang, Fangshan Dist., Beijing, 102488, China
| | - Jiechao Deng
- Department of Orthopedic Surgery, Liangxiang Hospital of Beijing Fangshan District, and Liangxiang Teaching Hospital of Capital Medical University, No.45, Gongchen Ave., Liangxiang, Fangshan Dist., Beijing, 102488, China
| | - Zishun Gong
- Department of Orthopedic Surgery, Liangxiang Hospital of Beijing Fangshan District, and Liangxiang Teaching Hospital of Capital Medical University, No.45, Gongchen Ave., Liangxiang, Fangshan Dist., Beijing, 102488, China
| |
Collapse
|
31
|
Jin R, Cui Y, Chen H, Zhang Z, Weng T, Xia S, Yu M, Zhang W, Shao J, Yang M, Han C, Wang X. Three-dimensional bioprinting of a full-thickness functional skin model using acellular dermal matrix and gelatin methacrylamide bioink. Acta Biomater 2021; 131:248-261. [PMID: 34265473 DOI: 10.1016/j.actbio.2021.07.012] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 06/27/2021] [Accepted: 07/04/2021] [Indexed: 11/29/2022]
Abstract
Treatment of full-thickness skin defects still presents a significant challenge in clinical practice. Three-dimensional (3D) bioprinting technique offers a promising approach for fabricating skin substitutes. However, it is necessary to identify bioinks that have both sufficient mechanical properties and desirable biocompatibilities. In this study, we successfully fabricated acellular dermal matrix (ADM) and gelatin methacrylamide (GelMA) bioinks. The results demonstrated that ADM preserved the main extracellular matrix (ECM) components of the skin and GelMA had tunable mechanical properties. Both bioinks with shear-thinning properties were suitable for 3D bioprinting and GelMA bioink exhibited high printability. Additionally, the results revealed that 20% GelMA with sufficient mechanical properties was suitable to engineer epidermis, 1.5% ADM and 10% GelMA displayed relatively good cytocompatibilities. Here, we proposed a new 3D structure to simulate natural full-thickness skin, which included 20% GelMA with HaCaTs as an epidermal layer, 1.5% ADM with fibroblasts as the dermis, and 10% GelMA mesh with human umbilical vein endothelial cells (HUVECs) as the vascular network and framework. We demonstrated that this 3D bioprinting functional skin model (FSM) could not only promote cell viability and proliferation, but also support epidermis reconstruction in vitro. When transplanted in vivo, the FSM could maintain cell viability for at least 1 week. Furthermore, the FSM promoted wound healing and re-epithelization, stimulated dermal ECM secretion and angiogenesis, and improved wound healing quality. The FSM may provide viable functional skin substitutes for future clinical applications. STATEMENT OF SIGNIFICANCE: We propose a new 3D structure to simulate natural full-thickness skin, which included 20% GelMA with HaCaTs as an epidermal layer, 1.5% ADM with fibroblasts as the dermis, and 10% GelMA mesh with HUVECs as the vascular network. It could not only maintain a moist microenvironment and barrier function, but also recreate the natural skin microenvironment to promote cell viability and proliferation. This may provide viable functional skin substitutes for future clinical applications.
Collapse
Affiliation(s)
- Ronghua Jin
- Department of Burns & Wound Care Center, The Second Affiliated Hospital of Zhejiang University College of Medicine, Hangzhou 310009, China
| | - Yuecheng Cui
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, China
| | - Haojiao Chen
- Department of Burns, Shaoxing Second Hospital, Shaoxing, China
| | - Zhenzhen Zhang
- First People's Hospital of Hangzhou Xiaoshan District, Hangzhou, China
| | - Tingting Weng
- Department of Burns & Wound Care Center, The Second Affiliated Hospital of Zhejiang University College of Medicine, Hangzhou 310009, China
| | - Sizhan Xia
- Department of Burns & Wound Care Center, The Second Affiliated Hospital of Zhejiang University College of Medicine, Hangzhou 310009, China
| | - Meirong Yu
- Clinical Research Center, The Second Affiliated Hospital of Zhejiang University College of Medicine, Hangzhou, China
| | - Wei Zhang
- Department of Burns & Wound Care Center, The Second Affiliated Hospital of Zhejiang University College of Medicine, Hangzhou 310009, China
| | - Jiaming Shao
- Department of Burns & Wound Care Center, The Second Affiliated Hospital of Zhejiang University College of Medicine, Hangzhou 310009, China
| | - Min Yang
- Department of Burns & Wound Care Center, The Second Affiliated Hospital of Zhejiang University College of Medicine, Hangzhou 310009, China
| | - Chunmao Han
- Department of Burns & Wound Care Center, The Second Affiliated Hospital of Zhejiang University College of Medicine, Hangzhou 310009, China
| | - Xingang Wang
- Department of Burns & Wound Care Center, The Second Affiliated Hospital of Zhejiang University College of Medicine, Hangzhou 310009, China.
| |
Collapse
|
32
|
Vascularization Strategies in Bone Tissue Engineering. Cells 2021; 10:cells10071749. [PMID: 34359919 PMCID: PMC8306064 DOI: 10.3390/cells10071749] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 07/05/2021] [Accepted: 07/06/2021] [Indexed: 12/12/2022] Open
Abstract
Bone is a highly vascularized tissue, and its development, maturation, remodeling, and regeneration are dependent on a tight regulation of blood vessel supply. This condition also has to be taken into consideration in the context of the development of artificial tissue substitutes. In classic tissue engineering, bone-forming cells such as primary osteoblasts or mesenchymal stem cells are introduced into suitable scaffolds and implanted in order to treat critical-size bone defects. However, such tissue substitutes are initially avascular. Because of the occurrence of hypoxic conditions, especially in larger tissue substitutes, this leads to the death of the implanted cells. Therefore, it is necessary to devise vascularization strategies aiming at fast and efficient vascularization of implanted artificial tissues. In this review article, we present and discuss the current vascularization strategies in bone tissue engineering. These are based on the use of angiogenic growth factors, the co-implantation of blood vessel forming cells, the ex vivo microfabrication of blood vessels by means of bioprinting, and surgical methods for creating surgically transferable composite tissues.
Collapse
|
33
|
Bouland C, Philippart P, Dequanter D, Corrillon F, Loeb I, Bron D, Lagneaux L, Meuleman N. Cross-Talk Between Mesenchymal Stromal Cells (MSCs) and Endothelial Progenitor Cells (EPCs) in Bone Regeneration. Front Cell Dev Biol 2021; 9:674084. [PMID: 34079804 PMCID: PMC8166285 DOI: 10.3389/fcell.2021.674084] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Accepted: 04/23/2021] [Indexed: 12/14/2022] Open
Abstract
Bone regeneration is a complex, well-orchestrated process based on the interactions between osteogenesis and angiogenesis, observed in both physiological and pathological situations. However, specific conditions (e.g., bone regeneration in large quantity, immunocompromised regenerative process) require additional support. Tissue engineering offers novel strategies. Bone regeneration requires a cell source, a matrix, growth factors and mechanical stimulation. Regenerative cells, endowed with proliferation and differentiation capacities, aim to recover, maintain, and improve bone functions. Vascularization is mandatory for bone formation, skeletal development, and different osseointegration processes. The latter delivers nutrients, growth factors, oxygen, minerals, etc. The development of mesenchymal stromal cells (MSCs) and endothelial progenitor cells (EPCs) cocultures has shown synergy between the two cell populations. The phenomena of osteogenesis and angiogenesis are intimately intertwined. Thus, cells of the endothelial line indirectly foster osteogenesis, and conversely, MSCs promote angiogenesis through different interaction mechanisms. In addition, various studies have highlighted the importance of the microenvironment via the release of extracellular vesicles (EVs). These EVs stimulate bone regeneration and angiogenesis. In this review, we describe (1) the phenomenon of bone regeneration by different sources of MSCs. We assess (2) the input of EPCs in coculture in bone regeneration and describe their contribution to the osteogenic potential of MSCs. We discuss (3) the interaction mechanisms between MSCs and EPCs in the context of osteogenesis: direct or indirect contact, production of growth factors, and the importance of the microenvironment via the release of EVs.
Collapse
Affiliation(s)
- Cyril Bouland
- Department of Stomatology and Maxillofacial Surgery, Saint-Pierre Hospital, Brussels, Belgium.,Laboratory of Clinical Cell Therapy, Jules Bordet Institute, Université Libre de Bruxelles, Brussels, Belgium
| | - Pierre Philippart
- Faculty of Medicine, Université Libre de Bruxelles, Brussels, Belgium.,Department of Stomatology and Maxillofacial Surgery, IRIS South Hospital, Brussels, Belgium
| | - Didier Dequanter
- Department of Stomatology and Maxillofacial Surgery, Saint-Pierre Hospital, Brussels, Belgium.,Faculty of Medicine, Université Libre de Bruxelles, Brussels, Belgium
| | - Florent Corrillon
- Laboratory of Clinical Cell Therapy, Jules Bordet Institute, Université Libre de Bruxelles, Brussels, Belgium
| | - Isabelle Loeb
- Department of Stomatology and Maxillofacial Surgery, Saint-Pierre Hospital, Brussels, Belgium.,Faculty of Medicine, Université Libre de Bruxelles, Brussels, Belgium
| | - Dominique Bron
- Faculty of Medicine, Université Libre de Bruxelles, Brussels, Belgium.,Department of Hematology, Jules Bordet Institute, Université Libre de Bruxelles, Brussels, Belgium
| | - Laurence Lagneaux
- Laboratory of Clinical Cell Therapy, Jules Bordet Institute, Université Libre de Bruxelles, Brussels, Belgium
| | - Nathalie Meuleman
- Laboratory of Clinical Cell Therapy, Jules Bordet Institute, Université Libre de Bruxelles, Brussels, Belgium.,Faculty of Medicine, Université Libre de Bruxelles, Brussels, Belgium.,Department of Hematology, Jules Bordet Institute, Université Libre de Bruxelles, Brussels, Belgium
| |
Collapse
|
34
|
Kawecki F, Galbraith T, Clafshenkel WP, Fortin M, Auger FA, Fradette J. In Vitro Prevascularization of Self-Assembled Human Bone-Like Tissues and Preclinical Assessment Using a Rat Calvarial Bone Defect Model. MATERIALS 2021; 14:ma14082023. [PMID: 33920607 PMCID: PMC8073395 DOI: 10.3390/ma14082023] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Revised: 04/14/2021] [Accepted: 04/15/2021] [Indexed: 12/15/2022]
Abstract
In vitro prevascularization has the potential to address the challenge of maintaining cell viability at the core of engineered constructs, such as bone substitutes, and to improve the survival of tissue grafts by allowing quicker anastomosis to the host microvasculature. The self-assembly approach of tissue engineering allows the production of biomimetic bone-like tissue constructs including extracellular matrix and living human adipose-derived stromal/stem cells (hASCs) induced towards osteogenic differentiation. We hypothesized that the addition of endothelial cells could improve osteogenesis and biomineralization during the production of self-assembled human bone-like tissues using hASCs. Additionally, we postulated that these prevascularized constructs would consequently improve graft survival and bone repair of rat calvarial bone defects. This study shows that a dense capillary network spontaneously formed in vitro during tissue biofabrication after two weeks of maturation. Despite reductions in osteocalcin levels and hydroxyapatite formation in vitro in prevascularized bone-like tissues (35 days of culture), in vivo imaging of prevascularized constructs showed an improvement in cell survival without impeding bone healing after 12 weeks of implantation in a calvarial bone defect model (immunocompromised male rats), compared to their stromal counterparts. Globally, these findings establish our ability to engineer prevascularized bone-like tissues with improved functional properties.
Collapse
Affiliation(s)
- Fabien Kawecki
- Centre de Recherche en Organogénèse Expérimentale de l′Université Laval/LOEX, Division of Regenerative Medicine, CHU de Québec Research Center-Université Laval, Québec, QC G1J 1Z4, Canada; (F.K.); (T.G.); (W.P.C.); (M.F.); (F.A.A.)
- Department of Surgery, Faculty of Medicine, Université Laval, Québec, QC G1V 0A6, Canada
| | - Todd Galbraith
- Centre de Recherche en Organogénèse Expérimentale de l′Université Laval/LOEX, Division of Regenerative Medicine, CHU de Québec Research Center-Université Laval, Québec, QC G1J 1Z4, Canada; (F.K.); (T.G.); (W.P.C.); (M.F.); (F.A.A.)
| | - William P. Clafshenkel
- Centre de Recherche en Organogénèse Expérimentale de l′Université Laval/LOEX, Division of Regenerative Medicine, CHU de Québec Research Center-Université Laval, Québec, QC G1J 1Z4, Canada; (F.K.); (T.G.); (W.P.C.); (M.F.); (F.A.A.)
| | - Michel Fortin
- Centre de Recherche en Organogénèse Expérimentale de l′Université Laval/LOEX, Division of Regenerative Medicine, CHU de Québec Research Center-Université Laval, Québec, QC G1J 1Z4, Canada; (F.K.); (T.G.); (W.P.C.); (M.F.); (F.A.A.)
- Faculty of Dentistry, Université Laval, Québec, QC G1V 0A6, Canada
- Service of Oral and Maxillofacial Surgery, CHU de Québec-Université Laval, Québec, QC G1J 1Z4, Canada
| | - François A. Auger
- Centre de Recherche en Organogénèse Expérimentale de l′Université Laval/LOEX, Division of Regenerative Medicine, CHU de Québec Research Center-Université Laval, Québec, QC G1J 1Z4, Canada; (F.K.); (T.G.); (W.P.C.); (M.F.); (F.A.A.)
- Department of Surgery, Faculty of Medicine, Université Laval, Québec, QC G1V 0A6, Canada
| | - Julie Fradette
- Centre de Recherche en Organogénèse Expérimentale de l′Université Laval/LOEX, Division of Regenerative Medicine, CHU de Québec Research Center-Université Laval, Québec, QC G1J 1Z4, Canada; (F.K.); (T.G.); (W.P.C.); (M.F.); (F.A.A.)
- Department of Surgery, Faculty of Medicine, Université Laval, Québec, QC G1V 0A6, Canada
- Correspondence:
| |
Collapse
|
35
|
Teotia AK, Qayoom I, Singh P, Mishra A, Jaiman D, Seppälä J, Lidgren L, Kumar A. Exosome-Functionalized Ceramic Bone Substitute Promotes Critical-Sized Bone Defect Repair in Rats. ACS APPLIED BIO MATERIALS 2021; 4:3716-3726. [DOI: 10.1021/acsabm.1c00311] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Arun K. Teotia
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur 201806, India
| | - Irfan Qayoom
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur 201806, India
| | - Prerna Singh
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur 201806, India
- Center for Environmental Science and Engineering, Indian Institute of Technology Kanpur, Kanpur 201806, India
| | - Ankita Mishra
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur 201806, India
| | - Deepika Jaiman
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur 201806, India
| | - Jukka Seppälä
- Polymer Technology, School of Chemical Engineering, Aalto University, Espoo 02150, Finland
| | - Lars Lidgren
- Department of Orthopedics, Clinical Sciences Lund, Lund University, Lund 221 85, Sweden
| | - Ashok Kumar
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur 201806, India
- Center for Environmental Science and Engineering, Indian Institute of Technology Kanpur, Kanpur 201806, India
- Center for Nanoscience, Indian Institute of Technology Kanpur, Kanpur 201806, India
- The Mehta Family Centre for Engineering in Medicine, Indian Institute of Technology Kanpur, Kanpur 208016, India
| |
Collapse
|
36
|
Wang X, Mei L, Jin M, Jiang X, Li X, Li J, Xu Y, Meng Z, Zhu J, Wu F. Composite Coating of Graphene Oxide/TiO2 Nanotubes/HHC-36 Antibacterial Peptide Construction and an Exploration of Its Bacteriostat and Osteogenesis Effects. J Biomed Nanotechnol 2021; 17:662-676. [PMID: 35057892 DOI: 10.1166/jbn.2021.3013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Graphene oxide (GO), a kind of polymer, is often selected as a controlled released agent, whereas titanium dioxide (TiO2) nanotubes are commonly used as a drug-coated carrier. This study was conducted to develop methods for manufacturing the GO/TiO2/HHC-36 composite
coating and exploring its bacteriostat and osteogenesis properties. The GO/TiO2 nanotubes were prepared by electrochemical methods and HHC-36 was then adsorbed to GO/TiO2to obtain GO/TiO2/HHC-36. Sustained release of HHC-36 was analyzed and the antibacterial
effect was examined by the inhibition zone test. The biocompatibility and osteogenesis in vitro of GO/TiO2/HHC-36 were explored. Finally, the osteogenesic property of the composite coating was investigated in a rat femoral defect model in vivo. GO/TiO2/HHC-36
was successfully prepared and had good controlled released performance in vitro. The inhibit zone size of S. aureus was 2.1 mm and that of E. coli was 3.0 mm. GO/TiO2/HHC-36 showed good biocompatibility with mesenchymal stem cells (MSCs) and promoted their adhesion,
migration, and differentiation. In addition, the secretion of alkaline phosphatase, collagen, mineralized matrix and osteoblast-related nutrient factors of MSCs was increased after treatment with GO/TiO2/HHC-36. Furthermore, GO/TiO2/HHC-36 also stimulated endotheliocytes
to secrete VEGF, leading to angiogenesis. Finally, implantation of GO/TiO2/HHC-36 in the rat femur defect model resulted in MSC migration and increased expression of osteoblast related proteins. The composite coating with controlled released of HHC-36 showed distinct antibacterial
properties and promoted osteogenesis in vitro and in vivo.
Collapse
Affiliation(s)
- Xiaojun Wang
- Department of Orthopedics, Huzhou Traditional Chinese Medicine Hospital, Affiliated Hospital to Zhejiang Chinese Medical University, Huzhou 313000, P. R.China
| | - Lina Mei
- Department of Internal Medicine, Huzhou Maternity & Child Health Care Hospital, Huzhou 313000, P. R. China
| | - Mingchao Jin
- Department of Orthopedics, Huzhou Central Hospital, Affiliated Central Hospital of Huzhou University, Huzhou Hospital of Zhejiang University, Huzhou 313000, P. R. China
| | - Xuesheng Jiang
- Department of Orthopedics, Huzhou Central Hospital, Affiliated Central Hospital of Huzhou University, Huzhou Hospital of Zhejiang University, Huzhou 313000, P. R. China
| | - Xiongfeng Li
- Department of Orthopedics, Huzhou Central Hospital, Affiliated Central Hospital of Huzhou University, Huzhou Hospital of Zhejiang University, Huzhou 313000, P. R. China
| | - Jianyou Li
- Department of Orthopedics, Huzhou Central Hospital, Affiliated Central Hospital of Huzhou University, Huzhou Hospital of Zhejiang University, Huzhou 313000, P. R. China
| | - Yan Xu
- Department of Rehabilitation, Huzhou Central Hospital, Affiliated Central Hospital of Huzhou University, Huzhou Hospital of Zhejiang University, Huzhou 313000, P. R. China
| | - Zhipeng Meng
- Department of Anesthesiology, Huzhou Central Hospital, Affiliated Central Hospital of Huzhou University, Huzhou Hospital of Zhejiang University, Huzhou 313000, P. R. China
| | - Junkun Zhu
- Orthopedics Rehabilitation Department, Lishui Municipal Central Hospital, Lishui 323000, P. R. China
| | - Fengfeng Wu
- Department of Orthopedics, Huzhou Central Hospital, Affiliated Central Hospital of Huzhou University, Huzhou Hospital of Zhejiang University, Huzhou 313000, P. R. China
- Department of Rehabilitation, Huzhou Central Hospital, Affiliated Central Hospital of Huzhou University, Huzhou Hospital of Zhejiang
University, Huzhou 313000, P. R. China
| |
Collapse
|
37
|
Vascular Endothelial Growth Factor and Mesenchymal Stem Cells Revealed Similar Bone Formation to Allograft in a Sheep Model. BIOMED RESEARCH INTERNATIONAL 2021; 2021:6676609. [PMID: 33763484 PMCID: PMC7946458 DOI: 10.1155/2021/6676609] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 02/07/2021] [Accepted: 02/25/2021] [Indexed: 01/08/2023]
Abstract
Introduction Mesenchymal stem cells (MSCs) and vascular endothelial growth factor (VEGF) are key factors in bone regeneration. Further stimulation should establish an enhanced cell environment optimal for vessel evolvement and hereby being able to attract bone-forming cells. The aim of this study was to generate new bone by using MSCs and VEGF, being able to stimulate growth equal to allograft. Methods Eight Texel/Gotland sheep had four titanium implants in a size of 10 × 12 mm inserted into bilateral distal femurs, containing a 2 mm gap. In the gap, autologous 3 × 106 MSCs seeded on hydroxyapatite (HA) granules in combination with 10 ng, 100 ng, and 500 ng VEGF release/day were added. After 12 weeks, the implant-bone blocks were harvested, embedded, and sectioned for histomorphometric analysis. Bone formation and mechanical fixation were evaluated. Blood samples were collected for the determination of bone-related biomarkers and VEGF in serum at weeks 0, 1, 4, 8, and 12. Results The combination of 3 × 106 MSCs with 10 ng, 100 ng, and 500 ng VEGF release/day exhibited similar amount of bone formation within the gap as allograft (P > 0.05). Moreover, no difference in mechanical fixation was observed between the groups (P > 0.05). Serum biomarkers showed no significant difference compared to baseline (all P > 0.05). Conclusion MSCs and VEGF exhibit significant bone regeneration, and their bone properties equal to allograft, with no systemic increase in osteogenic markers or VEGF with no visible side effects. This study indicates a possible new approach into solving the problem of insufficient allograft, in larger bone defects.
Collapse
|
38
|
Uusitalo-Kylmälä L, Santo Mendes AC, Polari L, Joensuu K, Heino TJ. An In Vitro Co-Culture Model of Bone Marrow Mesenchymal Stromal Cells and Peripheral Blood Mononuclear Cells Promotes the Differentiation of Myeloid Angiogenic Cells and Pericyte-Like Cells. Stem Cells Dev 2021; 30:309-324. [PMID: 33499756 DOI: 10.1089/scd.2019.0171] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Mesenchymal stromal cells (MSCs) are known to stimulate the survival and growth of endothelial cells (ECs) by producing paracrine signals, as well as to differentiate into pericytes and thereby support blood vessel formation and stability. On the other hand, cells with an EC-like phenotype have been found within the CD14+ and CD34+ cell populations of peripheral blood (PB) mononuclear cells (MNCs). The aim of this study was to investigate the proangiogenic differentiation potential of human MSC-MNC co-cultures. Bone marrow-derived MSCs (2,500 cells/cm2) were co-cultured with MNCs (50,000 cells/cm2), which were isolated from the PB of healthy donors. MSCs and MNCs cultured alone at same cell densities were used as controls. Cells in MNC fraction and in co-cultures were isolated for CD14, CD34, and CD31 surface markers with magnetic-activated cell sorting. Co-cultures were analyzed for cell proliferation and morphology, as well as for the expression of various hematopoietic, endothelial, and pericyte markers by immunocytochemistry, quantitative PCR (qPCR), and flow cytometry. Vascular endothelial growth factor (VEGF) expression and secretion was measured with qPCR and enzyme-linked immunosorbent assay, respectively. Our results show that in co-cultures with MSCs, CD14+CD45+ MNCs differentiated into spindle-shaped, nonproliferative, EC-like, myeloid angiogenic cells (MACs) expressing CD31, but also into pericyte-like cells expressing neural/glial antigen 2 (NG2) and CD146. Functionality of the isolated MACs was demonstrated in co-cultures with human umbilical vein endothelial cells, where they supported the formation of tube-like structures. NG2+ cells of MNC-origin were found among both CD34-CD14+ and CD34-CD14- cell populations, indicating the existence of different subtypes of pericyte-like cells. In addition, VEGF was shown to be secreted in MSC-MNC co-cultures, mainly by MSCs. In conclusion, MSCs were shown to possess proangiogenic capacity in MSC-MNC co-cultures as they supported the differentiation of functional MACs, as well as the differentiation of pericyte-like cells of MNC origin. This phenomenon was mediated at least partially via secreted VEGF.
Collapse
Affiliation(s)
| | - Ana Carolina Santo Mendes
- Institute of Biomedicine, University of Turku, Turku, Finland.,Department of Life Sciences, Faculty of Science and Technology, University of Coimbra, Coimbra, Portugal
| | - Lauri Polari
- Department of Biosciences, Cell Biology, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland
| | - Katriina Joensuu
- Institute of Biomedicine, University of Turku, Turku, Finland.,Department of Plastic Surgery, Tampere University Hospital, Tampere, Finland
| | - Terhi J Heino
- Institute of Biomedicine, University of Turku, Turku, Finland
| |
Collapse
|
39
|
Ma M, Yang W, Cai Z, Wang P, Li H, Mi R, Jiang Y, Xie Z, Sui P, Wu Y, Shen H. SMAD-specific E3 ubiquitin ligase 2 promotes angiogenesis by facilitating PTX3 degradation in MSCs from patients with ankylosing spondylitis. STEM CELLS (DAYTON, OHIO) 2021; 39:581-599. [PMID: 33547700 PMCID: PMC8248389 DOI: 10.1002/stem.3332] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 11/18/2020] [Accepted: 12/10/2020] [Indexed: 12/13/2022]
Abstract
Dysregulated angiogenesis of mesenchymal stem cells (MSCs) is closely related to inflammation and disrupted bone metabolism in patients with various autoimmune diseases. However, the role of MSCs in the development of abnormal angiogenesis in patients with ankylosing spondylitis (AS) remains unclear. In this study, we cultured human umbilical vein endothelial cells (HUVECs) with bone marrow-derived MSCs from patients with AS (ASMSCs) or healthy donors (HDMSCs) in vitro. Then, the cocultured HUVECs were assayed using a cell counting kit-8 (CCK-8) to evaluate the cell proliferation. A wound healing assay was performed to investigate cell migration, and a tube formation assay was conducted to determine the angiogenesis efficiency. ASMSCs exhibited increased angiogenesis, and increased expression of SMAD-specific E3 ubiquitin ligase 2 (Smurf2) in MSCs was the main cause of abnormal angiogenesis in patients with AS. Downregulation of Smurf2 in ASMSCs blocked angiogenesis, whereas overexpression of Smurf2 in HDMSCs promoted angiogenesis. The pro-angiogenic effect of Smurf2 was confirmed by the results of a Matrigel plug assay in vivo. By functioning as an E3 ubiquitin ligase in MSCs, Smurf2 regulated the levels of pentraxin 3 (PTX3), which has been shown to suppress angiogenesis through the PTX3-fibroblast growth factor 2 pathway. Moreover, Smurf2 transcription was regulated by activating transcription factor 4-induced endoplasmic reticulum stress. In conclusion, these results identify novel roles of Smurf2 in negatively regulating PTX3 stability and promoting angiogenesis in ASMSCs.
Collapse
Affiliation(s)
- Mengjun Ma
- Department of Orthopedics, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, People's Republic of China
| | - Wen Yang
- Department of Orthopedics, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, People's Republic of China
| | - Zhaopeng Cai
- Department of Orthopedics, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, People's Republic of China
| | - Peng Wang
- Department of Orthopedics, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, People's Republic of China
| | - Hongyu Li
- Department of Orthopedics, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, People's Republic of China
| | - Rujia Mi
- Center for Biotherapy, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, People's Republic of China
| | - Yuhang Jiang
- Department of Orthopedics, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, People's Republic of China
| | - Zhongyu Xie
- Department of Orthopedics, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, People's Republic of China
| | - Pengfei Sui
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, People's Republic of China
| | - Yanfeng Wu
- Center for Biotherapy, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, People's Republic of China
| | - Huiyong Shen
- Department of Orthopedics, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, People's Republic of China.,Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China
| |
Collapse
|
40
|
Adipose and Muscle Cell Co-Culture System: A Novel In Vitro Tool to Mimic the In Vivo Cellular Environment. BIOLOGY 2020; 10:biology10010006. [PMID: 33374127 PMCID: PMC7823969 DOI: 10.3390/biology10010006] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 12/14/2020] [Accepted: 12/23/2020] [Indexed: 12/18/2022]
Abstract
A co-culture system allows researchers to investigate the complex interactions between two cell types under various environments, such as those that promote differentiation and growth as well as those that mimic healthy and diseased states, in vitro. In this paper, we review the most common co-culture systems for myocytes and adipocytes. The in vitro techniques mimic the in vivo environment and are used to investigate the causal relationships between different cell lines. Here, we briefly discuss mono-culture and co-culture cell systems and their applicability to the study of communication between two or more cell types, including adipocytes and myocytes. Also, we provide details about the different types of co-culture systems and their applicability to the study of metabolic disease, drug development, and the role of secretory factors in cell signaling cascades. Therefore, this review provides details about the co-culture systems used to study the complex interactions between adipose and muscle cells in various environments, such as those that promote cell differentiation and growth and those used for drug development.
Collapse
|
41
|
Pereira I, Pereira JE, Maltez L, Rodrigues A, Rodrigues C, Oliveira M, Silva DM, Caseiro AR, Prada J, Maurício AC, Santos JD, Gama M. Regeneration of critical-sized defects, in a goat model, using a dextrin-based hydrogel associated with granular synthetic bone substitute. Regen Biomater 2020; 8:rbaa036. [PMID: 33732486 PMCID: PMC7947577 DOI: 10.1093/rb/rbaa036] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 08/03/2020] [Accepted: 08/06/2020] [Indexed: 12/27/2022] Open
Abstract
The development of injectable bone substitutes (IBS) have obtained great importance in the bone regeneration field, as a strategy to reach hardly accessible defects using minimally invasive techniques and able to fit to irregular topographies. In this scenario, the association of injectable hydrogels and bone graft granules is emerging as a well-established trend. Particularly, in situ forming hydrogels have arisen as a new IBS generation. An in situ forming and injectable dextrin-based hydrogel (HG) was developed, aiming to act as a carrier of granular bone substitutes and bioactive agents. In this work, the HG was associated to a granular bone substitute (Bonelike®) and implanted in goat critical-sized calvarial defects (14 mm) for 3, 6 and 12 weeks. The results showed that HG improved the handling properties of the Bonelike® granules and did not affect its osteoconductive features, neither impairing the bone regeneration process. Human multipotent mesenchymal stromal cells from the umbilical cord, extracellular matrix hydrolysates and the pro-angiogenic peptide LLKKK18 were also combined with the IBS. These bioactive agents did not enhance the new bone formation significantly under the conditions tested, according to micro-computed tomography and histological analysis.
Collapse
Affiliation(s)
- Isabel Pereira
- CEB, Centre of Biological Engineering, University of Minho, Campus de Gualtar, Braga 4710-057, Portugal
- Correspondence address. CEB, Centre of Biological Engineering, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal. Tel: +351-253-604-418; E-mail:
| | - José Eduardo Pereira
- CECAV, Animal and Veterinary Research Centre, University of Trás-os-Montes e Alto Douro, Vila Real 5001-801, Portugal
- Department of Veterinary Sciences, University of Trás-os-Montes e Alto Douro, Vila Real 5001-801, Portugal
| | - Luís Maltez
- CECAV, Animal and Veterinary Research Centre, University of Trás-os-Montes e Alto Douro, Vila Real 5001-801, Portugal
- Department of Veterinary Sciences, University of Trás-os-Montes e Alto Douro, Vila Real 5001-801, Portugal
| | - Alexandra Rodrigues
- CEB, Centre of Biological Engineering, University of Minho, Campus de Gualtar, Braga 4710-057, Portugal
| | - Catarina Rodrigues
- CEB, Centre of Biological Engineering, University of Minho, Campus de Gualtar, Braga 4710-057, Portugal
| | - Manuela Oliveira
- CEB, Centre of Biological Engineering, University of Minho, Campus de Gualtar, Braga 4710-057, Portugal
| | - Dina M Silva
- Biosckin, Molecular and Cell Therapies S.A., Laboratório Criovida, TecMaia, Rua Engenheiro Frederico Ulrich 2650, Moreira da Maia 4470-605, Portugal
| | - Ana Rita Caseiro
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, n° 228, Porto 4050-313, Portugal
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado 55142, Porto 4051-401 Portugal
- Centro de Investigação Vasco da Gama (CIVG)/Escola Universitária Vasco da Gama (EUVG), Avenida José R. Sousa Fernandes, n.° 197 Lordemão, Coimbra 3020-210, Portugal
| | - Justina Prada
- CECAV, Animal and Veterinary Research Centre, University of Trás-os-Montes e Alto Douro, Vila Real 5001-801, Portugal
- Department of Veterinary Sciences, University of Trás-os-Montes e Alto Douro, Vila Real 5001-801, Portugal
| | - Ana Colette Maurício
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, n° 228, Porto 4050-313, Portugal
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado 55142, Porto 4051-401 Portugal
| | - José Domingos Santos
- REQUIMTE/LAQV, Departamento de Engenharia Metalúrgica e Materiais, Faculdade de Engenharia, Universidade do Porto, Rua Dr Roberto Frias, Porto 4200-495, Portugal
| | - Miguel Gama
- CEB, Centre of Biological Engineering, University of Minho, Campus de Gualtar, Braga 4710-057, Portugal
| |
Collapse
|
42
|
Silk fibroin as a natural polymeric based bio-material for tissue engineering and drug delivery systems-A review. Int J Biol Macromol 2020; 163:2145-2161. [DOI: 10.1016/j.ijbiomac.2020.09.057] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 09/06/2020] [Accepted: 09/09/2020] [Indexed: 12/13/2022]
|
43
|
Leroux A, Paiva Dos Santos B, Leng J, Oliveira H, Amédée J. Sensory neurons from dorsal root ganglia regulate endothelial cell function in extracellular matrix remodelling. Cell Commun Signal 2020; 18:162. [PMID: 33076927 PMCID: PMC7574530 DOI: 10.1186/s12964-020-00656-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 09/06/2020] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Recent physiological and experimental data highlight the role of the sensory nervous system in bone repair, but its precise role on angiogenesis in a bone regeneration context is still unknown. Our previous work demonstrated that sensory neurons (SNs) induce the osteoblastic differentiation of mesenchymal stem cells, but the influence of SNs on endothelial cells (ECs) was not studied. METHODS Here, in order to study in vitro the interplay between SNs and ECs, we used microfluidic devices as an indirect co-culture model. Gene expression analysis of angiogenic markers, as well as measurements of metalloproteinases protein levels and enzymatic activity, were performed. RESULTS We were able to demonstrate that two sensory neuropeptides, calcitonin gene-related peptide (CGRP) and substance P (SP), were involved in the transcriptional upregulation of angiogenic markers (vascular endothelial growth factor, angiopoietin 1, type 4 collagen, matrix metalloproteinase 2) in ECs. Co-cultures of ECs with SNs also increased the protein level and enzymatic activity of matrix metalloproteinases 2 and 9 (MMP2/MMP9) in ECs. CONCLUSIONS Our results suggest a role of sensory neurons, and more specifically of CGRP and SP, in the remodelling of endothelial cells extracellular matrix, thus supporting and enhancing the angiogenesis process. Video abstract.
Collapse
Affiliation(s)
- Alice Leroux
- Univ. Bordeaux, INSERM, BIOTIS, U1026, F-33000, Bordeaux, France.
| | | | - Jacques Leng
- Univ. Bordeaux, CNRS, Solvay, LOF, UMR 5258, F-33006, Pessac, France
| | - Hugo Oliveira
- Univ. Bordeaux, INSERM, BIOTIS, U1026, F-33000, Bordeaux, France
| | - Joëlle Amédée
- Univ. Bordeaux, INSERM, BIOTIS, U1026, F-33000, Bordeaux, France
| |
Collapse
|
44
|
Heo DN, Ayan B, Dey M, Banerjee D, Wee H, Lewis GS, Ozbolat IT. Aspiration-assisted bioprinting of co-cultured osteogenic spheroids for bone tissue engineering. Biofabrication 2020; 13. [PMID: 33059343 DOI: 10.1088/1758-5090/abc1bf] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Accepted: 10/15/2020] [Indexed: 12/18/2022]
Abstract
Conventional top-down approaches in tissue engineering involving cell seeding on scaffolds have been widely used in bone engineering applications. However, scaffold-based bone tissue constructs have had limited clinical translation due to constrains in supporting scaffolds, minimal flexibility in tuning scaffold degradation, and low achievable cell seeding density as compared with native bone tissue. Here, we demonstrate a pragmatic and scalable bottom-up method, inspired from embryonic developmental biology, to build three-dimensional (3D) scaffold-free constructs using spheroids as building blocks. Human umbilical vein endothelial cells (HUVECs) were introduced to human mesenchymal stem cells (hMSCs) (hMSC/HUVEC) and spheroids were fabricated by an aggregate culture system. Bone tissue was generated by induction of osteogenic differentiation in hMSC/HUVEC spheroids for 10 days, with enhanced osteogenic differentiation and cell viability in the core of the spheroids compared to hMSC-only spheroids. Aspiration-assisted bioprinting (AAB) is a new bioprinting technique which allows precise positioning of spheroids (11% with respect to the spheroid diameter) by employing aspiration to lift individual spheroids and bioprint them onto a hydrogel. AAB facilitated bioprinting of scaffold-free bone tissue constructs using the pre-differentiated hMSC/HUVEC spheroids. These constructs demonstrated negligible changes in their shape for two days after bioprinting owing to the reduced proliferative potential of differentiated stem cells. Bioprinted bone tissues showed interconnectivity with actin-filament formation and high expression of osteogenic and endothelial-specific gene factors. This study thus presents a viable approach for 3D bioprinting of complex-shaped geometries using spheroids as building blocks, which can be used for various applications including but not limited to, tissue engineering, organ-on-a-chip and microfluidic devices, drug screening and, disease modeling.
Collapse
Affiliation(s)
| | - Bugra Ayan
- Penn State, University Park, Pennsylvania, UNITED STATES
| | - Madhuri Dey
- Penn State, University Park, Pennsylvania, UNITED STATES
| | | | - Hwabok Wee
- Penn State, Hershey, Pennsylvania, UNITED STATES
| | | | | |
Collapse
|
45
|
Liu R, Zhang S, Chen X. Injectable hydrogels for tendon and ligament tissue engineering. J Tissue Eng Regen Med 2020; 14:1333-1348. [PMID: 32495524 DOI: 10.1002/term.3078] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 05/06/2020] [Accepted: 05/17/2020] [Indexed: 01/14/2023]
Abstract
The problem of tendon and ligament (T/L) regeneration in musculoskeletal diseases has long constituted a major challenge. In situ injection of formable biodegradable hydrogels, however, has been demonstrated to treat T/L injury and reduce patient suffering in a minimally invasive manner. An injectable hydrogel is more suitable than other biological materials due to the special physiological structure of T/L. Most other materials utilized to repair T/L are cell-based, growth factor-based materials, with few material properties. In addition, the mechanical property of the gel cannot reach the normal T/L level. This review summarizes advances in natural and synthetic polymeric injectable hydrogels for tissue engineering in T/L and presents prospects for injectable and biodegradable hydrogels for its treatment. In future T/L applications, it is necessary develop an injectable hydrogel with mechanics, tissue damage-specific binding, and disease response. Simultaneously, the advantages of various biological materials must be combined in order to achieve personalized precision therapy.
Collapse
Affiliation(s)
- Richun Liu
- Guangxi Collaborative Innovation Center for Biomedicine, Guangxi Medical University, Nanning, Guangxi, China
| | - Shichen Zhang
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xiao Chen
- Guangxi Collaborative Innovation Center for Biomedicine, Guangxi Medical University, Nanning, Guangxi, China.,Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
46
|
Guerreiro SG, Unger RE, Cerqueira NMFSA, Sartoris A, Martins MJ, Barbosa MA, Soares R, Granja PL, Kirkpatrick CJ. Alkaline phosphatase dual-binding sites for collagen dictate cell migration and microvessel assembly in vitro. J Cell Biochem 2020; 122:116-129. [PMID: 32748513 DOI: 10.1002/jcb.29835] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 07/15/2020] [Accepted: 07/24/2020] [Indexed: 01/01/2023]
Abstract
Interactions between cell types, growth factors, and extracellular matrix components involved in angiogenesis are crucial for new vessel formation leading to tissue regeneration. This study investigated whether cocultures of fibroblasts and endothelial cells (ECs; from macro- or microvasculature) play a role in the formation of microvessel-like structures by ECs, as well as modulate fibroblast differentiation and growth factors production (vascular endothelial cell growth factor, basic fibroblast growth factor, active transforming growth factor-β1, and interleukin-8), which are important for vessel sprouting and maturation. Data obtained revealed that in vitro coculture systems of fibroblasts and human ECs stimulate collagen synthesis and growth factors production by fibroblasts that ultimately affect the formation and distribution of microvessel-like structures in cell cultures. In this study, areas with activated fibroblasts and high alkaline phosphatase (ALP) activity were also observed in cocultures. Molecular docking assays revealed that ALP has two binding positions for collagen, suggesting its impact in collagen proteins' aggregation, cell migration, and microvessel assembly. These findings indicate that bioinformatics and coculture systems are complementary tools for investigating the participation of proteins, like collagen and ALP in angiogenesis.
Collapse
Affiliation(s)
- Susana G Guerreiro
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal.,Instituto de Engenharia Biomédica (INEB), Universidade do Porto, Porto, Portugal.,Faculdade de Engenharia da Universidade do Porto (FEUP), Porto, Portugal.,Departamento de Biomedicina, Faculdade de Medicina da Universidade do Porto (FMUP), Porto, Portugal
| | - Ronald E Unger
- REPAIR-Lab, Institute of Pathology, University Medical Center, Johannes Gutenberg University, Mainz, Germany
| | - Nuno M F S A Cerqueira
- Departamento de Química e Bioquímica, UCIBIO@REQUIMTE, Faculdade de Ciências da Universidade do Porto, Porto, Portugal
| | - Anne Sartoris
- REPAIR-Lab, Institute of Pathology, University Medical Center, Johannes Gutenberg University, Mainz, Germany
| | - Maria J Martins
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal.,Departamento de Biomedicina, Faculdade de Medicina da Universidade do Porto (FMUP), Porto, Portugal
| | - Mário A Barbosa
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal.,Instituto de Engenharia Biomédica (INEB), Universidade do Porto, Porto, Portugal.,Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal
| | - Raquel Soares
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal.,Departamento de Biomedicina, Faculdade de Medicina da Universidade do Porto (FMUP), Porto, Portugal
| | - Pedro L Granja
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal.,Instituto de Engenharia Biomédica (INEB), Universidade do Porto, Porto, Portugal.,Faculdade de Engenharia da Universidade do Porto (FEUP), Porto, Portugal.,Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal
| | - Charles J Kirkpatrick
- REPAIR-Lab, Institute of Pathology, University Medical Center, Johannes Gutenberg University, Mainz, Germany
| |
Collapse
|
47
|
Junka R, Yu X. Polymeric nanofibrous scaffolds laden with cell-derived extracellular matrix for bone regeneration. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 113:110981. [PMID: 32487395 PMCID: PMC7292471 DOI: 10.1016/j.msec.2020.110981] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 04/14/2020] [Accepted: 04/16/2020] [Indexed: 12/11/2022]
Abstract
Bone tissue engineering aims to alleviate the shortage of available autograft material and the biological/mechanical incompatibility of allografts through fabrication of bioactive synthetic bone graft substitutes. However, these substitute grafting materials have insufficient biological potency that limits their clinical efficacy in regenerating large defects. Extracellular matrix, a natural tissue scaffold laden with biochemical and structural cues regulating cell adhesion and tissue morphogenesis, may be a versatile supplement that can extend its biological functionality to synthetic grafts. Embedding decellularized extracellular matrix (dECM) into synthetic polymers offers a promising strategy to enhance cellular response to synthetic materials, mitigate physical and mechanical limitations of dECMs, and improve clinical utility of synthetic bone grafts. Enriched with dECM biochemical cues, synthetic polymers can be readily fabricated into complex biocomposite grafts that mimic bone structure and stimulate endogenous cells to regenerate bone. In this study, cell-derived dECMs from osteoblast and endothelial cells were incorporated into polycaprolactone (PCL) solutions for electrospinning dual-layer nanofibrous scaffolds with osteogenic and vascular cues. The study examined the bioactivity of dECM scaffolds in osteoblast cultures for cell number, mineral deposits, and osteogenic markers, as well as regeneration of cortical bone defect in a rat femur. Scaffolds with osteoblast dECM had a significantly robust osteoblast proliferation, Alizarin Red staining/concentration, and osteopontin-positive extracellular deposits. Implanted scaffolds increased bone growth in femoral defects, and constructs with both osteogenic and vascular cues significantly improved cortical width. These findings demonstrate the potential to fabricate tailored biomimetic grafts with dECM cues and fibrous architecture for bone applications.
Collapse
Affiliation(s)
- Radoslaw Junka
- Department Biomedical Engineering, Stevens Institute of Technology, Hoboken, NJ 07030, United States of America
| | - Xiaojun Yu
- Department Biomedical Engineering, Stevens Institute of Technology, Hoboken, NJ 07030, United States of America.
| |
Collapse
|
48
|
Grémare A, Aussel A, Bareille R, Paiva Dos Santos B, Amédée J, Thébaud NB, Le Nihouannen D. A Unique Triculture Model to Study Osteoblasts, Osteoclasts, and Endothelial Cells. Tissue Eng Part C Methods 2020; 25:421-432. [PMID: 31169074 DOI: 10.1089/ten.tec.2018.0301] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
IMPACT STATEMENT In this article, we first developed a new medium to culture together primary human osteoblastic, osteoclastic, and endothelial cells (ECs) chosen to represent the three major bone cell tissues. Indeed, no study has been conducted on primary human cells and on the phenotype/activity retention of these three primary human cell types. Thus, we established an original triculture model with osteoblastic, osteoclastic, and ECs, where not only both cell phenotype and cell activity were maintained but also cell culture homeostasis. These promising results will permit further investigations to create in vitro conditions to mimic the bone microenvironment and analyze cell interactions in ex vivo studies.
Collapse
Affiliation(s)
- Agathe Grémare
- 1Université de Bordeaux, INSERM, Tissue Bioengineering, U1026, CHU Bordeaux, Services d'Odontologie et de Santé Buccale, F-33076, Bordeaux, France
| | - Audrey Aussel
- 1Université de Bordeaux, INSERM, Tissue Bioengineering, U1026, CHU Bordeaux, Services d'Odontologie et de Santé Buccale, F-33076, Bordeaux, France
| | - Reine Bareille
- 2Université de Bordeaux, INSERM, Tissue Bioengineering, U1026, F-33076, Bordeaux, France
| | - Bruno Paiva Dos Santos
- 2Université de Bordeaux, INSERM, Tissue Bioengineering, U1026, F-33076, Bordeaux, France
| | - Joelle Amédée
- 2Université de Bordeaux, INSERM, Tissue Bioengineering, U1026, F-33076, Bordeaux, France
| | - Noélie B Thébaud
- 1Université de Bordeaux, INSERM, Tissue Bioengineering, U1026, CHU Bordeaux, Services d'Odontologie et de Santé Buccale, F-33076, Bordeaux, France
| | - Damien Le Nihouannen
- 2Université de Bordeaux, INSERM, Tissue Bioengineering, U1026, F-33076, Bordeaux, France
| |
Collapse
|
49
|
Han G, Zheng Z, Pan Z, Lin Y, Gan S, Jiao Y, Li H, Zhou C, Ding S, Li L. Sulfated chitosan coated polylactide membrane enhanced osteogenic and vascularization differentiation in MC3T3-E1s and HUVECs co-cultures system. Carbohydr Polym 2020; 245:116522. [PMID: 32718626 DOI: 10.1016/j.carbpol.2020.116522] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 05/24/2020] [Accepted: 05/25/2020] [Indexed: 12/22/2022]
Abstract
This study aimed to compare the effects of the two type chitosan derivatives, sulfated chitosan (SCS) and phosphorylated chitosan (PCS), coated on poly(d,l-lactide) (PDLLA) membrane via polydopamine, respectively, on vascularization and osteogenesis in vitro. Mouse preosteoblast cells (MC3T3-E1s) and human umbilical vein endothelial cells (HUVECs) were used as co-cultures system. The effects of two type membranes on calcium deposition, alkaline phosphatase (ALP) activity, vascularization related factors nitric oxide (NO) and angiogenic growth factor vascular endothelial growth factor (VEGF) were assessed. The changes of osteogenic and angiogenic related gene, and protein expression were evaluated too. In fact, SCS modified PDLLA membrane had the highest related gene and protein expression than other PDLLA membranes. Our results demonstrated that the SCS maybe a promising matrix for bone regeneration by co-cultures of ECs and OCs than PCS.
Collapse
Affiliation(s)
- Guijuan Han
- Department of Materials Science and Engineering, Jinan University, Guangzhou 510632, PR China
| | - Zexiang Zheng
- Department of Materials Science and Engineering, Jinan University, Guangzhou 510632, PR China
| | - Zhicheng Pan
- Department of Materials Science and Engineering, Jinan University, Guangzhou 510632, PR China
| | - Yucheng Lin
- Department of Materials Science and Engineering, Jinan University, Guangzhou 510632, PR China
| | - Shuchun Gan
- Department of Materials Science and Engineering, Jinan University, Guangzhou 510632, PR China
| | - Yanpeng Jiao
- Department of Materials Science and Engineering, Jinan University, Guangzhou 510632, PR China; Engineering Research Centre of Artificial Organs & Materials, Jinan University, Guangzhou 510632, PR China
| | - Hong Li
- Department of Materials Science and Engineering, Jinan University, Guangzhou 510632, PR China; Engineering Research Centre of Artificial Organs & Materials, Jinan University, Guangzhou 510632, PR China
| | - Changren Zhou
- Department of Materials Science and Engineering, Jinan University, Guangzhou 510632, PR China; Engineering Research Centre of Artificial Organs & Materials, Jinan University, Guangzhou 510632, PR China
| | - Shan Ding
- Department of Materials Science and Engineering, Jinan University, Guangzhou 510632, PR China; Engineering Research Centre of Artificial Organs & Materials, Jinan University, Guangzhou 510632, PR China.
| | - Lihua Li
- Department of Materials Science and Engineering, Jinan University, Guangzhou 510632, PR China; Engineering Research Centre of Artificial Organs & Materials, Jinan University, Guangzhou 510632, PR China.
| |
Collapse
|
50
|
Mutschall H, Winkler S, Weisbach V, Arkudas A, Horch RE, Steiner D. Bone tissue engineering using adipose-derived stem cells and endothelial cells: Effects of the cell ratio. J Cell Mol Med 2020; 24:7034-7043. [PMID: 32394620 PMCID: PMC7299704 DOI: 10.1111/jcmm.15374] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Revised: 04/06/2020] [Accepted: 04/22/2020] [Indexed: 12/29/2022] Open
Abstract
The microvascular endothelial network is essential for bone formation and regeneration. In this context, endothelial cells not only support vascularization but also influence bone physiology via cell contact-dependent mechanisms. In order to improve vascularization and osteogenesis in tissue engineering applications, several strategies have been developed. One promising approach is the coapplication of endothelial and adipose derived stem cells (ADSCs). In this study, we aimed at investigating the best ratio of human umbilical vein endothelial cells (HUVECs) and osteogenic differentiated ADSCs with regard to proliferation, apoptosis, osteogenesis and angiogenesis. For this purpose, cocultures of ADSCs and HUVECs with ratios of 25%:75%, 50%:50% and 75%:25% were performed. We were able to prove that cocultivation supports proliferation whereas apoptosis was unidirectional decreased in cocultured HUVECs mediated by a p-BAD-dependent mechanism. Moreover, coculturing ADSCs and HUVECs stimulated matrix mineralization and the activity of alkaline phosphatase (ALP). Increased gene expression of the proangiogenic markers eNOS, Flt, Ang2 and MMP3 as well as sprouting phenomena in matrigel assays proved the angiogenic potential of the coculture. In summary, coculturing ADSCs and HUVECs stimulates proliferation, cell survival, osteogenesis and angiogenesis particularly in the 50%:50% coculture.
Collapse
Affiliation(s)
- Hilkea Mutschall
- Department of Plastic and Hand Surgery, University Hospital of Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Sophie Winkler
- Department of Plastic and Hand Surgery, University Hospital of Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Volker Weisbach
- Department of Transfusion Medicine, University Hospital of Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Andreas Arkudas
- Department of Plastic and Hand Surgery, University Hospital of Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Raymund E Horch
- Department of Plastic and Hand Surgery, University Hospital of Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Dominik Steiner
- Department of Plastic and Hand Surgery, University Hospital of Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Erlangen, Germany
| |
Collapse
|