1
|
Li J, Zhou Y, Shu T, Lei W, Tang Q, Yang Y, Zhang J, Chen W, Zhou B, Hu Q, Xing Y, Wang J, Wang C. Differentiation of lung tissue-resident c-Kit + cells into microvascular endothelial cells alleviates pulmonary vascular remodeling. Dev Cell 2025:S1534-5807(25)00030-9. [PMID: 39909047 DOI: 10.1016/j.devcel.2025.01.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 10/28/2024] [Accepted: 01/15/2025] [Indexed: 02/07/2025]
Abstract
Pulmonary vascular remodeling (PVR), encompassing microvascular loss and muscularization, contributes to multiple respiratory diseases. c-Kit+ cells exhibit differentiation potential into both endothelial cells (ECs) and smooth muscle cells. The potential role of lung c-Kit+ cell differentiation in PVR, however, remains unclear. Lung c-Kit+ cells increase in pulmonary hypertension patients and in the SU5416/hypoxia (SuHx)-induced PVR mouse model. Employing genetic lineage tracing and single-cell RNA sequencing (scRNA-seq), we elucidate that lung-resident c-Kit+ cells display an aerocyte and venular endothelial differentiation in the SuHx model. Ablation of tissue-resident c-Kit+ cells exacerbates PVR. We identify an Nr2f2-expressing c-Kit+ cell subgroup, which exhibitsvenous EC differentiation and increases during PVR. Notably, the elevation of Nr2f2 in c-Kit+ cells via AAV enhances differentiation and mitigates PVR. These findings underscore the protective role of lung tissue-resident c-Kit+ cells in PVR, achieved by differentiating into mature ECs. Targeting NR2F2 expression in c-Kit+ cells emerges as a promising strategy for reversing PVR.
Collapse
Affiliation(s)
- Jinqiu Li
- State Key Laboratory of Respiratory Health and Multimorbidity, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing 100005, China
| | - Yitian Zhou
- Department of Physiology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100005, China
| | - Ting Shu
- State Key Laboratory of Respiratory Health and Multimorbidity, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing 100005, China; Haihe Laboratory of Cell Ecosystem, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing 100005, China
| | - Wenqi Lei
- State Key Laboratory of Respiratory Health and Multimorbidity, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing 100005, China
| | - Qihao Tang
- State Key Laboratory of Respiratory Health and Multimorbidity, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing 100005, China
| | - Yang Yang
- State Key Laboratory of Respiratory Health and Multimorbidity, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing 100005, China
| | - Jin Zhang
- Department of Thoracic Surgery, China-Japan Friendship Hospital, Beijing 100029, China
| | - Wenhui Chen
- Department of Lung Transplantation, China-Japan Friendship Hospital, Beijing 100029, China
| | - Bin Zhou
- New Cornerstone Investigator Institute, 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 200031, China
| | - Qinghua Hu
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yanjiang Xing
- State Key Laboratory of Respiratory Health and Multimorbidity, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing 100005, China; Haihe Laboratory of Cell Ecosystem, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing 100005, China.
| | - Jing Wang
- State Key Laboratory of Respiratory Health and Multimorbidity, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing 100005, China; Haihe Laboratory of Cell Ecosystem, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing 100005, China.
| | - Chen Wang
- State Key Laboratory of Respiratory Health and Multimorbidity, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing 100005, China; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, China-Japan Friendship Hospital, Beijing 100029, China
| |
Collapse
|
2
|
Duquesne J, Parmentier L, Vermeersch E, Lemaire F, Seo JW, Dmitriev RI, Vlierberghe SV. Volumetric bioprinting of the osteoid niche. Biofabrication 2025; 17:025002. [PMID: 39819878 DOI: 10.1088/1758-5090/adab25] [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: 01/04/2024] [Accepted: 01/16/2025] [Indexed: 01/19/2025]
Abstract
Volumetric bioprinting has revolutionized the field of biofabrication by enabling the creation of cubic centimeter-scale living constructs at faster printing times (in the order of seconds). However, a key challenge remains: developing a wider variety of available osteogenic bioinks that allow osteogenic maturation of the encapsulated cells within the construct. Herein, the bioink exploiting a step-growth mechanism (norbornene-norbornene functionalized gelatin in combination with thiolated gelatin-GelNBNBSH) outperformed the bioink exploiting a chain-growth mechanism (gelatin methacryloyl-GelMA), as the necessary photo-initiator concentration was three times lower combined with a more than 50% reduction in required light exposure dose resulting in an improved positive and negative resolution. To mimic the substrate elasticity of the osteoid, two concentrations of the photo-initiator Li-TPO-L (1 and 10 mg ml-1) were compared for post-curing whereby the lowest concentration was selected since it resulted in attaining the osteogenic substrate elasticity combined with excellent biocompatibility with HT1080 cells (>95%). Further physico-chemical testing revealed that the volumetric printing (VP) process affected the degradation time of the constructs with volumetric constructs degrading slower than the control sheets which could be due to the introduced fibrillar structure inherent to the VP process. Moreover, GelNBNBSH volumetric constructs significantly outperformed the GelMA volumetric constructs in terms of a 2-fold increase in photo-crosslinkable moiety conversion and a 3-fold increase in bulk stiffness of the construct. Finally, a 21-day osteogenic cell study was performed with highly viable dental pulp-derived stem cells (>95%) encapsulated within the volumetric printed constructs. Osteogenesis was greatly favored for the GelNBNBSH constructs through enhanced early (alkaline phosphatase activity) and late maturation (calcium production) osteogenic markers. After 21 d, a secretome analysis revealed a more mature osteogenic phenotype within GelNBNBSH constructs as compared to their chain-growth counterpart in terms of osteogenic, immunological and angiogenic signaling.
Collapse
Affiliation(s)
- Jessie Duquesne
- Polymer Chemistry and Biomaterials (PBM) Group, Centre of Macromolecular Chemistry (CMaC), Department of Organic and Macromolecular Chemistry, Faculty of Sciences, Ghent University, Krijgslaan 281, Building S4, 9000 Ghent, Belgium
| | - Laurens Parmentier
- Polymer Chemistry and Biomaterials (PBM) Group, Centre of Macromolecular Chemistry (CMaC), Department of Organic and Macromolecular Chemistry, Faculty of Sciences, Ghent University, Krijgslaan 281, Building S4, 9000 Ghent, Belgium
| | - Edward Vermeersch
- Polymer Chemistry and Biomaterials (PBM) Group, Centre of Macromolecular Chemistry (CMaC), Department of Organic and Macromolecular Chemistry, Faculty of Sciences, Ghent University, Krijgslaan 281, Building S4, 9000 Ghent, Belgium
| | - Flora Lemaire
- Biomatériaux et Inflammation en Site Osseux (BIOS), Université de Reims Champagne Ardenne, Avenue du Maréchal Juin 1, 51100 Reims, France
| | - Jung Won Seo
- Nano-biotechnology Laboratory, Department of Biotechnology, Faculty of Bioscience Engineering, Ghent university, Proeftuinstraat 86, 9000 Ghent, Belgium
| | - Ruslan I Dmitriev
- Tissue Engineering and Biomaterials Group, Department of Human Structure and Repair, Faculty of Medical and Health Sciences, Ghent University, C. Heymanslaan 10, 9000 Ghent, Belgium
| | - Sandra Van Vlierberghe
- Polymer Chemistry and Biomaterials (PBM) Group, Centre of Macromolecular Chemistry (CMaC), Department of Organic and Macromolecular Chemistry, Faculty of Sciences, Ghent University, Krijgslaan 281, Building S4, 9000 Ghent, Belgium
| |
Collapse
|
3
|
Campione P, Rizzo MG, Bauso LV, Ielo I, Messina GML, Calabrese G. Osteoblastic Differentiation of Human Adipose-Derived Mesenchymal Stem Cells on P3HT Thin Polymer Film. J Funct Biomater 2025; 16:10. [PMID: 39852566 PMCID: PMC11765816 DOI: 10.3390/jfb16010010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 12/20/2024] [Accepted: 12/31/2024] [Indexed: 01/26/2025] Open
Abstract
Bone defects restoration has always been an arduous challenge in the orthopedic field due to the limitations of conventional grafts. Bone tissue engineering offers an alternative approach by using biomimetic materials, stem cells, and growth factors that are able to improve the regeneration of bone tissue. Different biomaterials have attracted great interest in BTE applications, including the poly(3-hexylthiofene) (P3HT) conductive polymer, whose primary advantage is its capability to provide a native extracellular matrix-like environment. Based on this evidence, in this study, we evaluated the biological response of human adipose-derived mesenchymal stem cells cultured on P3HT thin polymer film for 14 days. Our results suggest that P3HT represents a good substrate to induce osteogenic differentiation of osteoprogenitor cells, even in the absence of specific inductive growth factors, thus representing a promising strategy for bone regenerative medicine. Therefore, the system provided may offer an innovative platform for next-generation biocompatible materials for regenerative medicine.
Collapse
Affiliation(s)
- Paola Campione
- Department of Chemical Sciences, University of Catania and CSGI, Viale A. Doria 6, 95125 Catania, Italy;
| | - Maria Giovanna Rizzo
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences (ChiBioFarAm), University of Messina, Viale F. Stagno d’Alcontres 31, 98166 Messina, Italy; (M.G.R.); (L.V.B.); (I.I.)
| | - Luana Vittoria Bauso
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences (ChiBioFarAm), University of Messina, Viale F. Stagno d’Alcontres 31, 98166 Messina, Italy; (M.G.R.); (L.V.B.); (I.I.)
| | - Ileana Ielo
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences (ChiBioFarAm), University of Messina, Viale F. Stagno d’Alcontres 31, 98166 Messina, Italy; (M.G.R.); (L.V.B.); (I.I.)
| | - Grazia Maria Lucia Messina
- Department of Chemical Sciences, University of Catania and CSGI, Viale A. Doria 6, 95125 Catania, Italy;
| | - Giovanna Calabrese
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences (ChiBioFarAm), University of Messina, Viale F. Stagno d’Alcontres 31, 98166 Messina, Italy; (M.G.R.); (L.V.B.); (I.I.)
| |
Collapse
|
4
|
Asakura T, Diep TTT, Ueda Y, Yamada A, Tsuzuno T, Takahashi N, Miyata M, Tabeta K, Nagata M, Matsuda K. Analysis of the Effect of Human Type I Collagen-Derived Peptide on Bone Regenerative Capacity and Comparison with Various Collagen Materials In Vivo. MEDICINA (KAUNAS, LITHUANIA) 2025; 61:57. [PMID: 39859038 PMCID: PMC11766864 DOI: 10.3390/medicina61010057] [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: 11/09/2024] [Revised: 12/11/2024] [Accepted: 12/23/2024] [Indexed: 01/27/2025]
Abstract
Background and Objectives: Autologous bone grafting is the first choice for reconstructive surgery in bone defects due to trauma or malignant tumors. However, there is an increasing demand for minimally invasive alternatives involving bone regeneration using artificial materials. Biomimetic materials that replicate the body's microscopic structure, such as Cellnest®, are gaining attention. Cellnest is a xeno-free recombinant peptide based on human type I collagen, containing a rich Arg-Gly-Asp (RGD) motif related to cell adhesion. The aim of this study was to compare the effects of Cellnest with existing collagen materials (Pelnac®, Integra®, Terudermis®) on bone regeneration and elucidate the underlying mechanisms. Materials and Methods: In vivo experiments involved a rat model of calvarial bone defects, in which Cellnest and other collagen materials were implanted into the defect area. Bone formation was assessed after 4 weeks using micro-computed tomography (micro-CT) and histological analysis. In vitro experiments included the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT), adhesion, and migration assays, and a real-time polymerase chain reaction using rapidly expanding cells (RECs) to explore the mechanisms of Cellnest's bone regenerative capacity. Results: The micro-CT analysis showed that the regenerated bone area was significantly greater in the Cellnest group (72.3%) than in the Pelnac® (25.5%), Integra® (31.6%), and Terudermis® (38.3%) groups. The histological analysis confirmed similar trends, with Cellnest showing 42.2% bone regeneration, outperforming the other materials. The in vitro assays revealed that Cellnest promoted cell proliferation, adhesion, and migration. Gene expression analysis demonstrated that Cellnest significantly increased the levels of the bone formation markers ALP and COL1. Conclusions: Cellnest, a human type I collagen-like peptide rich in RGD motifs, enhances bone regeneration by promoting MSC adhesion and migration, and bone formation-related gene expression. The findings suggest its potential as an effective material for bone defect reconstruction.
Collapse
Affiliation(s)
- Tatsunori Asakura
- Department of Plastic and Reconstructive Surgery, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8510, Japan
| | - Tran Thi Thuy Diep
- Division of Periodontology, Department of Oral Biological Science, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8514, Japan
| | - Yuta Ueda
- Division of Periodontology, Department of Oral Biological Science, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8514, Japan
| | - Aoi Yamada
- Division of Pioneering Advanced Therapeutics, Niigata University Medical and Dental Hospital, Niigata 951-8520, Japan
| | - Takahiro Tsuzuno
- Division of Periodontology, Department of Oral Biological Science, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8514, Japan
| | - Naoki Takahashi
- Division of Periodontology, Department of Oral Biological Science, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8514, Japan
| | - Masayuki Miyata
- Department of Plastic and Reconstructive Surgery, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8510, Japan
| | - Koichi Tabeta
- Division of Periodontology, Department of Oral Biological Science, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8514, Japan
| | - Masaki Nagata
- Division of Pioneering Advanced Therapeutics, Niigata University Medical and Dental Hospital, Niigata 951-8520, Japan
| | - Ken Matsuda
- Department of Plastic and Reconstructive Surgery, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8510, Japan
| |
Collapse
|
5
|
Zhang Y, Zhang J, Lesani P, Lu Z, Zreiqat H. Osteopontin Rejuvenates Senescent Adipose-Derived Stem Cells and Restores their Bone Tissue Regenerative Function. Stem Cell Rev Rep 2024; 20:1106-1120. [PMID: 38472643 PMCID: PMC11087332 DOI: 10.1007/s12015-024-10707-5] [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] [Accepted: 02/26/2024] [Indexed: 03/14/2024]
Abstract
The regenerative function of stem cells is compromised when the proportion of senescent stem cells increases with ageing advance. Therefore, combating stem cell senescence is of great importance for stem cell-based tissue engineering in the elderly, but remains largely unexplored. Osteopontin (OPN), a glycosylated phosphoprotein, is one of the key extracellular matrix molecules in bone tissue. OPN activates various signalling pathways and modulates cellular activities, including cell senescence. However, the role of OPN in stem cell senescence remains largely unknown. This study aims to investigate if OPN modulates cell senescence and bone regenerative function in human adipose-derived mesenchymal stem cells (ASCs), and to determine the underlying mechanisms. We first developed a senescent ASC model using serial passaging until passage 10 (P10), in which senescent cells were characterised by reduced proliferation and osteogenic differentiation capacity compared to P4 ASCs. The conditioned medium from P10 ASCs exhibited a diminished trophic effect on human osteoblasts (HOBs), compared to that from P4 ASCs. P10 ASCs on OPN-coated surface showed rejuvenated phenotype and enhanced osteogenic differentiation. The conditioned medium from P10 ASCs on OPN-coating improved trophic effects on HOBs. OPN regulated the morphology of senescent ASCs, transforming them from a more rounded and flattened cell shape to an elongated shape with a smaller area. These findings demonstrated the effects of OPN in restoring senescent ASCs functions, possibly through a mechanism that involves the modulation of cell morphology, indicating that OPN might hold a great potential for rejuvenating senescent stem cells and could potentially open a new venue for regenerating bone tissue in age-related diseases.
Collapse
Affiliation(s)
- Yiran Zhang
- Tissue Engineering & Biomaterials Research Unit, School of Biomedical Engineering, Faculty of Engineering and IT, The University of Sydney, Darlington, NSW, 2006, Australia
| | - Junni Zhang
- Tissue Engineering & Biomaterials Research Unit, School of Biomedical Engineering, Faculty of Engineering and IT, The University of Sydney, Darlington, NSW, 2006, Australia
| | - Pooria Lesani
- Tissue Engineering & Biomaterials Research Unit, School of Biomedical Engineering, Faculty of Engineering and IT, The University of Sydney, Darlington, NSW, 2006, Australia
| | - Zufu Lu
- Tissue Engineering & Biomaterials Research Unit, School of Biomedical Engineering, Faculty of Engineering and IT, The University of Sydney, Darlington, NSW, 2006, Australia.
| | - Hala Zreiqat
- Tissue Engineering & Biomaterials Research Unit, School of Biomedical Engineering, Faculty of Engineering and IT, The University of Sydney, Darlington, NSW, 2006, Australia.
| |
Collapse
|
6
|
Azadi S, Yazdanpanah MA, Afshari A, Alahdad N, Chegeni S, Angaji A, Rezayat SM, Tavakol S. Bioinspired synthetic peptide-based biomaterials regenerate bone through biomimicking of extracellular matrix. J Tissue Eng 2024; 15:20417314241303818. [PMID: 39670180 PMCID: PMC11635874 DOI: 10.1177/20417314241303818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Accepted: 11/15/2024] [Indexed: 12/14/2024] Open
Abstract
There have been remarkable advancements in regenerative medicine for bone regeneration, tackling the worldwide health concern of tissue loss. Tissue engineering uses the body's natural capabilities and applies biomaterials and bioactive molecules to replace damaged or lost tissues and restore their functionality. While synthetic ceramics have overcome some challenges associated with allografts and xenografts, they still need essential growth factors and biomolecules. Combining ceramics and bioactive molecules, such as peptides derived from biological motifs of vital proteins, is the most effective approach to achieve optimal bone regeneration. These bioactive peptides induce various cellular processes and modify scaffold properties by mimicking the function of natural osteogenic, angiogenic and antibacterial biomolecules. The present review aims to consolidate the latest and most pertinent information on the advancements in bioactive peptides, including angiogenic, osteogenic, antimicrobial, and self-assembling peptide nanofibers for bone tissue regeneration, elucidating their biological effects and potential clinical implications.
Collapse
Affiliation(s)
- Sareh Azadi
- Department of Medical Biotechnology, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Ali Yazdanpanah
- Department of Cell and Molecular Biology, Faculty of Biological Science, Kharazmi University, Tehran, Iran
| | - Ali Afshari
- Department of Cell and Molecular Biology, Faculty of Biological Science, Kharazmi University, Tehran, Iran
| | - Niloofar Alahdad
- Department of Cell and Molecular Biology, Faculty of Biological Science, Kharazmi University, Tehran, Iran
| | - Solmaz Chegeni
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Abdolhamid Angaji
- Department of Medical Biotechnology, Iran University of Medical Sciences, Tehran, Iran
| | - Seyed Mahdi Rezayat
- Department of Medical Nanotechnology, Tehran University of Medical Sciences, Tehran, Iran
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Shima Tavakol
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Research and Development, Tavakol Biomimetic Technologies Company, Tehran, Iran
| |
Collapse
|
7
|
Kechagias S, Theodoridis K, Broomfield J, Malpartida-Cardenas K, Reid R, Georgiou P, van Arkel RJ, Jeffers JRT. The effect of nodal connectivity and strut density within stochastic titanium scaffolds on osteogenesis. Front Bioeng Biotechnol 2023; 11:1305936. [PMID: 38107615 PMCID: PMC10721980 DOI: 10.3389/fbioe.2023.1305936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Accepted: 11/20/2023] [Indexed: 12/19/2023] Open
Abstract
Modern orthopaedic implants use lattice structures that act as 3D scaffolds to enhance bone growth into and around implants. Stochastic scaffolds are of particular interest as they mimic the architecture of trabecular bone and can combine isotropic properties and adjustable structure. The existing research mainly concentrates on controlling the mechanical and biological performance of periodic lattices by adjusting pore size and shape. Still, less is known on how we can control the performance of stochastic lattices through their design parameters: nodal connectivity, strut density and strut thickness. To elucidate this, four lattice structures were evaluated with varied strut densities and connectivity, hence different local geometry and mechanical properties: low apparent modulus, high apparent modulus, and two with near-identical modulus. Pre-osteoblast murine cells were seeded on scaffolds and cultured in vitro for 28 days. Cell adhesion, proliferation and differentiation were evaluated. Additionally, the expression levels of key osteogenic biomarkers were used to assess the effect of each design parameter on the quality of newly formed tissue. The main finding was that increasing connectivity increased the rate of osteoblast maturation, tissue formation and mineralisation. In detail, doubling the connectivity, over fixed strut density, increased collagen type-I by 140%, increased osteopontin by 130% and osteocalcin by 110%. This was attributed to the increased number of acute angles formed by the numerous connected struts, which facilitated the organization of cells and accelerated the cell cycle. Overall, increasing connectivity and adjusting strut density is a novel technique to design stochastic structures which combine a broad range of biomimetic properties and rapid ossification.
Collapse
Affiliation(s)
- Stylianos Kechagias
- Department of Mechanical Engineering, Imperial College London, London, United Kingdom
| | | | - Joseph Broomfield
- Centre for Bio Inspired Technology, Department of Electrical and Electronic Engineering, Imperial College London, London, United Kingdom
- Department of Surgery and Cancer, Imperial College London, London, United Kingdom
| | - Kenny Malpartida-Cardenas
- Centre for Bio Inspired Technology, Department of Electrical and Electronic Engineering, Imperial College London, London, United Kingdom
- Department of Infectious Disease, Imperial College London, London, United Kingdom
| | - Ruth Reid
- Centre for Bio Inspired Technology, Department of Electrical and Electronic Engineering, Imperial College London, London, United Kingdom
| | - Pantelis Georgiou
- Centre for Bio Inspired Technology, Department of Electrical and Electronic Engineering, Imperial College London, London, United Kingdom
| | - Richard J. van Arkel
- Department of Mechanical Engineering, Imperial College London, London, United Kingdom
| | | |
Collapse
|
8
|
Liang Y, Gao W, Deng S, Wu D, Jiang Y, Zhang Y, Qiu R. Graphene quantum dots promote migration and differentiation of periodontal ligament stem cells. Front Chem 2023; 11:1213507. [PMID: 38025053 PMCID: PMC10679356 DOI: 10.3389/fchem.2023.1213507] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 11/01/2023] [Indexed: 12/01/2023] Open
Abstract
Graphene and its derivatives have attracted much attention as nanomaterials in bone tissue engineering because of their remarkable ability to induce cell osteogenic differentiation. However, graphene quantum dots (GQDs), as graphene derivatives, little is known about their osteodifferentiation- and osteoinduction-promoting capabilities, especially in the restoration of bone defect caused by periodontitis. Therefore, there is a growing need to investigate the effect of GQDs on periodontal ligament stem cells (PDLSCs). Here, we postulated that GQDs are a promising biocompatible nanomaterial that facilitated the migration and differentiation of PDLSCs, and use laboratory methods like CCK-8, transwell experiments, qRT-PCR, Alizarin red staining and immunofluorescence staining to evaluate. Our experiments confirmed that GQDs did not inhibit cell viability, with most cells remaining viable even at GQDs concentrations of up to 30 μg mL-1. Moreover, GQDs were found to significantly enhance PDLSC migration, with the peak effect observed at concentrations of 5 and 10 μg mL-1. Furthermore, GQDs accelerated osteoblastic differentiation in PDLSCs and induced the mineralization of calcium nodules. Additionally, GQDs were shown to promote fibroblast differentiation in PDLSCs compared to the control group. Thus, GQDs not only possessed low cytotoxicity and good biocompatibility, but also displayed the beneficial capability to migration and differentiation of PDLSCs, which indicated GQDs might be a potential nanomaterial for bone regeneration.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Rongmin Qiu
- Guangxi Key Laboratory of Oral and Maxillofacial Surgery Disease Treatment, Guangxi Health Commission Key Laboratory of Prevention and Treatment for Oral Infectious Diseases, Guangxi Key Laboratory of Oral and Maxillofacial Rehabilitation and Reconstruction, Guangxi Clinical Research Center for Craniofacial Deformity, College of Stomatology, Hospital of Stomatology, Guangxi Medical University, Nanning, China
| |
Collapse
|
9
|
Jaber M, Hofbauer LC, Hofbauer C, Duda GN, Checa S. Reduced Bone Regeneration in Rats With Type 2 Diabetes Mellitus as a Result of Impaired Stromal Cell and Osteoblast Function-A Computer Modeling Study. JBMR Plus 2023; 7:e10809. [PMID: 38025037 PMCID: PMC10652174 DOI: 10.1002/jbm4.10809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 07/18/2023] [Accepted: 08/02/2023] [Indexed: 12/01/2023] Open
Abstract
Bone has the fascinating ability to self-regenerate. However, under certain conditions, such as type 2 diabetes mellitus (T2DM), this ability is impaired. T2DM is a chronic metabolic disease known by the presence of elevated blood glucose levels that is associated with reduced bone regeneration capability, high fracture risk, and eventual non-union risk after a fracture. Several mechanical and biological factors relevant to bone regeneration have been shown to be affected in a diabetic environment. However, whether impaired bone regeneration in T2DM can be explained due to mechanical or biological alterations remains unknown. To elucidate the relevance of either one, the aim of this study was to investigate the relative contribution of T2DM-related alterations on either cellular activity or mechanical stimuli driving bone regeneration. A previously validated in silico computer modeling approach that was capable of explaining bone regeneration in uneventful conditions of healing was further developed to investigate bone regeneration in T2DM. Aspects analyzed included the presence of mesenchymal stromal cells (MSCs), cellular migration, proliferation, differentiation, apoptosis, and cellular mechanosensitivity. To further verify the computer model findings against in vivo data, an experimental setup was replicated, in which regeneration was compared in healthy and diabetic after a rat femur bone osteotomy stabilized with plate fixation. We found that mechanical alterations had little effect on the reduced bone regeneration in T2DM and that alterations in MSC proliferation, MSC migration, and osteoblast differentiation had the highest effect. In silico predictions of regenerated bone in T2DM matched qualitatively and quantitatively those from ex vivo μCT at 12 weeks post-surgery when reduced cellular activities reported in previous in vitro and in vivo studies were included in the model. The presented findings here could have clinical implications in the treatment of bone fractures in patients with T2DM. © 2023 The Authors. JBMR Plus published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Mahdi Jaber
- Julius Wolff Institute at Berlin Institute of Health, Charité—Universitätsmedizin BerlinBerlinGermany
| | - Lorenz C Hofbauer
- Department of Medicine III and Center for Healthy AgingTechnische Universität DresdenDresdenGermany
| | - Christine Hofbauer
- Department of Medicine III and Center for Healthy AgingTechnische Universität DresdenDresdenGermany
| | - Georg N Duda
- Julius Wolff Institute at Berlin Institute of Health, Charité—Universitätsmedizin BerlinBerlinGermany
- BIH Center for Regenerative TherapiesBIH at Charité ‐ Universitätsmedizin BerlinBerlinGermany
| | - Sara Checa
- Julius Wolff Institute at Berlin Institute of Health, Charité—Universitätsmedizin BerlinBerlinGermany
| |
Collapse
|
10
|
Zhang Y, He F, Zhang Q, Lu H, Yan S, Shi X. 3D-Printed Flat-Bone-Mimetic Bioceramic Scaffolds for Cranial Restoration. RESEARCH (WASHINGTON, D.C.) 2023; 6:0255. [PMID: 37899773 PMCID: PMC10603392 DOI: 10.34133/research.0255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 10/04/2023] [Indexed: 10/31/2023]
Abstract
The limitations of autologous bone grafts necessitate the development of advanced biomimetic biomaterials for efficient cranial defect restoration. The cranial bones are typical flat bones with sandwich structures, consisting of a diploe in the middle region and 2 outer compact tables. In this study, we originally developed 2 types of flat-bone-mimetic β-tricalcium phosphate bioceramic scaffolds (Gyr-Comp and Gyr-Tub) by high-precision vat-photopolymerization-based 3-dimensional printing. Both scaffolds had 2 outer layers and an inner layer with gyroid pores mimicking the diploe structure. The outer layers of Gyr-Comp scaffolds simulated the low porosity of outer tables, while those of Gyr-Tub scaffolds mimicked the tubular pore structure in the tables of flat bones. The Gyr-Comp and Gyr-Tub scaffolds possessed higher compressive strength and noticeably promoted in vitro cell proliferation, osteogenic differentiation, and angiogenic activities compared with conventional scaffolds with cross-hatch structures. After implantation into rabbit cranial defects for 12 weeks, Gyr-Tub achieved the best repairing effects by accelerating the generation of bone tissues and blood vessels. This work provides an advanced strategy to prepare biomimetic biomaterials that fit the structural and functional needs of efficacious bone regeneration.
Collapse
Affiliation(s)
- Yihang Zhang
- School of Electromechanical Engineering,
Guangdong University of Technology, Guangzhou 510006, P. R. China
| | - Fupo He
- School of Electromechanical Engineering,
Guangdong University of Technology, Guangzhou 510006, P. R. China
| | - Qiang Zhang
- School of Materials Science and Engineering,
South China University of Technology, Guangzhou 510641, P. R. China
| | - Haotian Lu
- Peking Union Medical College Graduate School, Beijing 100730, P. R. China
| | - Shengtao Yan
- Peking Union Medical College Graduate School, Beijing 100730, P. R. China
- Department of Emergency,
China-Japan Friendship Hospital, Beijing 100029, P. R. China
| | - Xuetao Shi
- School of Materials Science and Engineering,
South China University of Technology, Guangzhou 510641, P. R. China
| |
Collapse
|
11
|
Cárdenas-Aguazaco W, Camacho B, Gómez-Pachón EY, Lara-Bertrand AL, Silva-Cote I. Electrospun Scaffolds of Polylactic Acid, Collagen, and Amorphous Calcium Phosphate for Bone Repair. Pharmaceutics 2023; 15:2529. [PMID: 38004509 PMCID: PMC10674189 DOI: 10.3390/pharmaceutics15112529] [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: 08/31/2023] [Revised: 10/14/2023] [Accepted: 10/16/2023] [Indexed: 11/26/2023] Open
Abstract
Most electrospun scaffolds for bone tissue engineering typically use hydroxyapatite (HA) or beta tricalcium phosphate (β-TCP). However, the biological activity of these crystalline compounds can be limited due to their low solubility. Therefore, amorphous calcium phosphate (ACP) may be an alternative in bone repair scaffolds. This study analyzes the morphology, porosity, mechanical strength, and surface chemistry of electrospun scaffolds composed of polylactic acid and collagen integrated with hydroxyapatite (MHAP) or amorphous calcium phosphate (MACP). In addition, the in vitro biocompatibility, osteogenic differentiation, and growth factor production associated with bone repair using human Wharton's jelly-derived mesenchymal stem cells (hWJ-MSCs) are evaluated. The results show that the electrospun MHAP and MACP scaffolds exhibit a fibrous morphology with interconnected pores. Both scaffolds exhibit favorable biocompatibility and stimulate the proliferation and osteogenesis of hWJ-MSCs. However, cell adhesion and osteocalcin production are greater in the MACP scaffold compared to the MHAP scaffold. In addition, the MACP scaffold shows significant production of bone-repair-related growth factors such as transforming growth factor-beta 1 (TGF-β1), providing a solid basis for its use in bone tissue engineering.
Collapse
Affiliation(s)
- William Cárdenas-Aguazaco
- Instituto Distrital de Ciencia, Biotecnología e Innovación en Salud-IDCBIS, Bogotá 111611, Colombia; (W.C.-A.); (B.C.); (A.L.L.-B.)
| | - Bernardo Camacho
- Instituto Distrital de Ciencia, Biotecnología e Innovación en Salud-IDCBIS, Bogotá 111611, Colombia; (W.C.-A.); (B.C.); (A.L.L.-B.)
| | - Edwin Yesid Gómez-Pachón
- Facultad Duitama, Universidad Pedagógica y Tecnológica de Colombia-UPTC, Duitama 150462, Colombia;
| | - Adriana Lorena Lara-Bertrand
- Instituto Distrital de Ciencia, Biotecnología e Innovación en Salud-IDCBIS, Bogotá 111611, Colombia; (W.C.-A.); (B.C.); (A.L.L.-B.)
| | - Ingrid Silva-Cote
- Instituto Distrital de Ciencia, Biotecnología e Innovación en Salud-IDCBIS, Bogotá 111611, Colombia; (W.C.-A.); (B.C.); (A.L.L.-B.)
| |
Collapse
|
12
|
Ding Q, Zhang S, Liu X, Zhao Y, Yang J, Chai G, Wang N, Ma S, Liu W, Ding C. Hydrogel Tissue Bioengineered Scaffolds in Bone Repair: A Review. Molecules 2023; 28:7039. [PMID: 37894518 PMCID: PMC10609504 DOI: 10.3390/molecules28207039] [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: 09/07/2023] [Revised: 09/27/2023] [Accepted: 10/09/2023] [Indexed: 10/29/2023] Open
Abstract
Large bone defects due to trauma, infections, and tumors are difficult to heal spontaneously by the body's repair mechanisms and have become a major hindrance to people's daily lives and economic development. However, autologous and allogeneic bone grafts, with their lack of donors, more invasive surgery, immune rejection, and potential viral transmission, hinder the development of bone repair. Hydrogel tissue bioengineered scaffolds have gained widespread attention in the field of bone repair due to their good biocompatibility and three-dimensional network structure that facilitates cell adhesion and proliferation. In addition, loading natural products with nanoparticles and incorporating them into hydrogel tissue bioengineered scaffolds is one of the most effective strategies to promote bone repair due to the good bioactivity and limitations of natural products. Therefore, this paper presents a brief review of the application of hydrogels with different gel-forming properties, hydrogels with different matrices, and nanoparticle-loaded natural products loaded and incorporated into hydrogels for bone defect repair in recent years.
Collapse
Affiliation(s)
- Qiteng Ding
- College of Traditional Chinese Medicine, Jilin Agricultural University, Changchun 130118, China; (Q.D.); (S.Z.); (J.Y.); (S.M.)
| | - Shuai Zhang
- College of Traditional Chinese Medicine, Jilin Agricultural University, Changchun 130118, China; (Q.D.); (S.Z.); (J.Y.); (S.M.)
| | - Xinglong Liu
- College of Traditional Chinese Medicine, Jilin Agriculture Science and Technology College, Jilin 132101, China;
| | - Yingchun Zhao
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250117, China;
| | - Jiali Yang
- College of Traditional Chinese Medicine, Jilin Agricultural University, Changchun 130118, China; (Q.D.); (S.Z.); (J.Y.); (S.M.)
| | - Guodong Chai
- College of Resources and Environment, Jilin Agricultural University, Changchun 130118, China; (G.C.); (N.W.)
| | - Ning Wang
- College of Resources and Environment, Jilin Agricultural University, Changchun 130118, China; (G.C.); (N.W.)
| | - Shuang Ma
- College of Traditional Chinese Medicine, Jilin Agricultural University, Changchun 130118, China; (Q.D.); (S.Z.); (J.Y.); (S.M.)
| | - Wencong Liu
- School of Food and Pharmaceutical Engineering, Wuzhou University, Wuzhou 543002, China
| | - Chuanbo Ding
- College of Traditional Chinese Medicine, Jilin Agriculture Science and Technology College, Jilin 132101, China;
- Scientific and Technological Innovation Center of Health Products and Medical Materials with Characteristic Resources of Jilin Province, Changchun 130118, China
| |
Collapse
|
13
|
Szwed-Georgiou A, Płociński P, Kupikowska-Stobba B, Urbaniak MM, Rusek-Wala P, Szustakiewicz K, Piszko P, Krupa A, Biernat M, Gazińska M, Kasprzak M, Nawrotek K, Mira NP, Rudnicka K. Bioactive Materials for Bone Regeneration: Biomolecules and Delivery Systems. ACS Biomater Sci Eng 2023; 9:5222-5254. [PMID: 37585562 PMCID: PMC10498424 DOI: 10.1021/acsbiomaterials.3c00609] [Citation(s) in RCA: 53] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 07/31/2023] [Indexed: 08/18/2023]
Abstract
Novel tissue regeneration strategies are constantly being developed worldwide. Research on bone regeneration is noteworthy, as many promising new approaches have been documented with novel strategies currently under investigation. Innovative biomaterials that allow the coordinated and well-controlled repair of bone fractures and bone loss are being designed to reduce the need for autologous or allogeneic bone grafts eventually. The current engineering technologies permit the construction of synthetic, complex, biomimetic biomaterials with properties nearly as good as those of natural bone with good biocompatibility. To ensure that all these requirements meet, bioactive molecules are coupled to structural scaffolding constituents to form a final product with the desired physical, chemical, and biological properties. Bioactive molecules that have been used to promote bone regeneration include protein growth factors, peptides, amino acids, hormones, lipids, and flavonoids. Various strategies have been adapted to investigate the coupling of bioactive molecules with scaffolding materials to sustain activity and allow controlled release. The current manuscript is a thorough survey of the strategies that have been exploited for the delivery of biomolecules for bone regeneration purposes, from choosing the bioactive molecule to selecting the optimal strategy to synthesize the scaffold and assessing the advantages and disadvantages of various delivery strategies.
Collapse
Affiliation(s)
- Aleksandra Szwed-Georgiou
- Department
of Immunology and Infectious Biology, Faculty of Biology and Environmental
Protection, University of Lodz, Lodz 90-136, Poland
| | - Przemysław Płociński
- Department
of Immunology and Infectious Biology, Faculty of Biology and Environmental
Protection, University of Lodz, Lodz 90-136, Poland
| | - Barbara Kupikowska-Stobba
- Biomaterials
Research Group, Lukasiewicz Research Network
- Institute of Ceramics and Building Materials, Krakow 31-983, Poland
| | - Mateusz M. Urbaniak
- Department
of Immunology and Infectious Biology, Faculty of Biology and Environmental
Protection, University of Lodz, Lodz 90-136, Poland
- The
Bio-Med-Chem Doctoral School, University of Lodz and Lodz Institutes
of the Polish Academy of Sciences, University
of Lodz, Lodz 90-237, Poland
| | - Paulina Rusek-Wala
- Department
of Immunology and Infectious Biology, Faculty of Biology and Environmental
Protection, University of Lodz, Lodz 90-136, Poland
- The
Bio-Med-Chem Doctoral School, University of Lodz and Lodz Institutes
of the Polish Academy of Sciences, University
of Lodz, Lodz 90-237, Poland
| | - Konrad Szustakiewicz
- Department
of Polymer Engineering and Technology, Faculty of Chemistry, Wroclaw University of Technology, Wroclaw 50-370, Poland
| | - Paweł Piszko
- Department
of Polymer Engineering and Technology, Faculty of Chemistry, Wroclaw University of Technology, Wroclaw 50-370, Poland
| | - Agnieszka Krupa
- Department
of Immunology and Infectious Biology, Faculty of Biology and Environmental
Protection, University of Lodz, Lodz 90-136, Poland
| | - Monika Biernat
- Biomaterials
Research Group, Lukasiewicz Research Network
- Institute of Ceramics and Building Materials, Krakow 31-983, Poland
| | - Małgorzata Gazińska
- Department
of Polymer Engineering and Technology, Faculty of Chemistry, Wroclaw University of Technology, Wroclaw 50-370, Poland
| | - Mirosław Kasprzak
- Biomaterials
Research Group, Lukasiewicz Research Network
- Institute of Ceramics and Building Materials, Krakow 31-983, Poland
| | - Katarzyna Nawrotek
- Faculty
of Process and Environmental Engineering, Lodz University of Technology, Lodz 90-924, Poland
| | - Nuno Pereira Mira
- iBB-Institute
for Bioengineering and Biosciences, Department of Bioengineering, Instituto Superior Técnico, Universidade de
Lisboa, Lisboa 1049-001, Portugal
- Associate
Laboratory i4HB-Institute for Health and Bioeconomy at Instituto Superior
Técnico, Universidade de Lisboa, Lisboa 1049-001, Portugal
- Instituto
Superior Técnico, Universidade de Lisboa, Lisboa 1049-001, Portugal
| | - Karolina Rudnicka
- Department
of Immunology and Infectious Biology, Faculty of Biology and Environmental
Protection, University of Lodz, Lodz 90-136, Poland
| |
Collapse
|
14
|
Alves L, Machado V, Botelho J, Mendes JJ, Cabral JMS, da Silva CL, Carvalho MS. Enhanced Proliferative and Osteogenic Potential of Periodontal Ligament Stromal Cells. Biomedicines 2023; 11:biomedicines11051352. [PMID: 37239023 DOI: 10.3390/biomedicines11051352] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 04/24/2023] [Accepted: 04/27/2023] [Indexed: 05/28/2023] Open
Abstract
Cell-based therapies using periodontal ligament stromal cells (PDLSC) for periodontal regeneration may represent an alternative source for mesenchymal stromal cells (MSC) to MSC derived from bone marrow (MSC(M)) and adipose tissue (MSC(AT)). We aimed to characterize the osteogenic/periodontal potential of PDLSC in comparison to MSC(M) and MSC(AT). PDLSC were obtained from surgically extracted healthy human third molars, while MSC(M) and MSC(AT) were obtained from a previously established cell bank. Flow cytometry, immunocytochemistry, and cell proliferation analyses provided cellular characteristics from each group. Cells from the three groups presented MSC-like morphology, MSC-related marker expression, and multilineage differentiation capacity (adipogenic, chondrogenic, and osteogenic). In this study, PDLSC expressed osteopontin, osteocalcin, and asporin, while MSC(M) and MSC(AT) did not. Of note, only PDLSC expressed CD146, a marker previously applied to identify PDLSC, and presented higher proliferative potential compared to MSC(M) and MSC(AT). Upon osteogenic induction, PDLSC exhibited higher calcium content and enhanced upregulation of osteogenic/periodontal genes compared to MSC(M) and MSC(AT), such as Runx2, Col1A1 and CEMP-1. However, the alkaline phosphatase activity of PDLSC did not increase. Our findings suggest that PDLSC might be a promising cell source for periodontal regeneration, presenting enhanced proliferative and osteogenic potential compared to MSC(M) and MSC(AT).
Collapse
Affiliation(s)
- Laura Alves
- Department of Bioengineering and iBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
| | - Vanessa Machado
- Clinical Research Unit, Egas Moniz Center for Interdisciplinary Research, Egas Moniz School of Health and Science, 2829-511 Almada, Portugal
- Evidence-Based Hub, Egas Moniz Center for Interdisciplinary Research, Egas Moniz School of Health and Science, 2829-511 Almada, Portugal
| | - João Botelho
- Clinical Research Unit, Egas Moniz Center for Interdisciplinary Research, Egas Moniz School of Health and Science, 2829-511 Almada, Portugal
- Evidence-Based Hub, Egas Moniz Center for Interdisciplinary Research, Egas Moniz School of Health and Science, 2829-511 Almada, Portugal
| | - José João Mendes
- Clinical Research Unit, Egas Moniz Center for Interdisciplinary Research, Egas Moniz School of Health and Science, 2829-511 Almada, Portugal
- Evidence-Based Hub, Egas Moniz Center for Interdisciplinary Research, Egas Moniz School of Health and Science, 2829-511 Almada, Portugal
| | - Joaquim M S Cabral
- Department of Bioengineering and iBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
| | - Cláudia L da Silva
- Department of Bioengineering and iBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
| | - Marta S Carvalho
- Department of Bioengineering and iBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
| |
Collapse
|
15
|
Methylphenidate Promotes Premature Growth Plate Closure: In Vitro Evidence. Int J Mol Sci 2023; 24:ijms24044175. [PMID: 36835608 PMCID: PMC9968202 DOI: 10.3390/ijms24044175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 01/25/2023] [Accepted: 01/27/2023] [Indexed: 02/22/2023] Open
Abstract
It is well known that patients with attention deficit hyperactivity disorder treated with stimulants, such as methylphenidate hydrochloride (MPH), have reduced height and weight. Even though MPH has an anorexigenic effect, an additional impact of this drug on the growth plate cannot be discarded. In this study, we aimed to determine the cellular effect of MPH on an in vitro growth plate model. We tested the effects of MPH on the viability and proliferation of a prechondrogenic cell line via an MTT assay. In vitro differentiation of this cell line was performed, and cell differentiation was evaluated through the expression of cartilage- and bone-related genes as measured via RT-PCR. MPH did not alter the viability or proliferation of prechondrogenic cells. However, it reduced the expression of cartilage extracellular matrix-related genes (type II collagen and aggrecan) and increased the expression of genes involved in growth plate calcification (Runx2, type I collagen, and osteocalcin) at different phases of their differentiation process. Our results evidence that MPH upregulates genes associated with growth plate hypertrophic differentiation. This may induce premature closure of the growth plate, which would contribute to the growth retardation that has been described to be induced by this drug.
Collapse
|
16
|
Barreiro BOB, Koth VS, Sesterheim P, Salum FG, Rübensam G, Augustin AH, Cherubini K. Autogenous dentin combined with mesenchymal stromal cells as an alternative alveolar bone graft: an in vivo study. Clin Oral Investig 2022; 27:1907-1922. [PMID: 36574044 DOI: 10.1007/s00784-022-04840-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 12/21/2022] [Indexed: 12/28/2022]
Abstract
OBJECTIVE Considering the chemical and structural properties of dentin, this study was aimed at evaluating the effect of dentin matrix alone or combined with mesenchymal stromal cells (MSC) on postextraction alveolar bone regeneration. MATERIAL AND METHODS Wistar rats were subjected to tooth extraction with osteotomy and allocated into groups according to the graft inserted: (1) Gelita-Spon®, (2) Bio-Oss®, (3) Dentin, (4) MSC, (5) Dentin/MSC, and (6) Control. Maxillae were analyzed by means of hematoxylin and eosin (H&E) staining, immunohistochemical (IHC) analysis, microcomputed tomography (micro-CT), and scanning electron microscopy (SEM). Serum levels of calcium and phosphorus were quantified. RESULTS The Bio-Oss group showed less bone than Gelita-Spon and Dentin/MSC; no other significant differences were seen in H&E analysis. The Bio-Oss group showed higher expression of collagen type I compared to the Dentin and Dentin/MSC groups and also higher osteocalcin expression than the Dentin/MSC group. There was a tendency of higher expression of osteopontin in the MSC, Dentin, and Dentin/MSC groups and higher VEGF in the MSC group. On micro-CT analysis, the Bio-Oss and the Dentin/MSC groups exhibited greater bone volume than the Control. Serum calcium and phosphorus levels did not significantly differ between the groups. SEM analysis depicted particles of Bio-Oss and dentin in the respective groups, as well as significant cellularity in the MSC group. CONCLUSION Autogenous nondemineralized dentin is an alternative for alveolar bone grafting, which can be improved by combination with MSC. CLINICAL RELEVANCE This work provides support for the clinical applicability of dentin graft alone or combined with MSC.
Collapse
Affiliation(s)
- Bernardo Ottoni Braga Barreiro
- School of Health and Life Sciences, Post-Graduate Program in Dentistry, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Valesca Sander Koth
- School of Health and Life Sciences, Post-Graduate Program in Dentistry, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Patrícia Sesterheim
- Experimental Cardiology Center, Institute of Cardiology of Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Fernanda Gonçalves Salum
- School of Health and Life Sciences, Post-Graduate Program in Dentistry, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Gabriel Rübensam
- Toxicology and Pharmacology Research Center (INTOX), School of Health and Life Sciences, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Adolpho Herbert Augustin
- Institute of Petroleum and Natural Resources (IPR), Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Karen Cherubini
- School of Health and Life Sciences, Post-Graduate Program in Dentistry, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, RS, Brazil.
- Serviço de Estomatologia, Hospital São Lucas PUCRS, Av. Ipiranga, 6690 Sala 231, Bairro Jardim Botânico, CEP: 90.610-000, Porto Alegre, RS, Brazil.
| |
Collapse
|
17
|
Iizumi R, Honda M. Wnt/β-Catenin Signaling Inhibits Osteogenic Differentiation in Human Periodontal Ligament Fibroblasts. Biomimetics (Basel) 2022; 7:biomimetics7040224. [PMID: 36546925 PMCID: PMC9776043 DOI: 10.3390/biomimetics7040224] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 11/23/2022] [Accepted: 12/01/2022] [Indexed: 12/07/2022] Open
Abstract
The periodontal ligament is a collagenous tissue that is important for maintaining the homeostasis of cementum and alveolar bone. In tendon cells, Wnt/β-catenin signaling has been reported to regulate the expression level of Scleraxis (Scx) and Mohawk Homeobox (Mkx) gene and maintain the tissue homeostasis, while its role in the periodontal ligament is unclear. The aim of this study was to investigate the effects of Wnt/β-catenin signaling induced by Wnt-3a stimulation on the inhibition of osteogenic differentiation of human periodontal ligament fibroblasts (HPLFs). During osteogenic differentiation of HPLFs, they formed bone nodules independently of alkaline phosphatase (ALP) activity. After stimulation of Wnt-3a, the expression of β-catenin increased, and nuclear translocation of β-catenin was observed. These data indicate that Wnt-3a activated Wnt/β-catenin signaling. Furthermore, the stimulation of Wnt-3a inhibited the bone nodule formation and suppressed the expression of osteogenic differentiation-related genes such as Runx2, Osteopontin and Osteocalcin, and upregulated the gene expression of Type-I collagen and Periostin (Postn). Scx may be involved in the suppression of osteogenic differentiation in HPLFs. In conclusion, Wnt/β-catenin signaling may be an important signaling pathway that inhibits the osteogenic differentiation in HPLFs by the upregulation of Scx gene expression and downregulation of osteogenic differentiation-related genes.
Collapse
|
18
|
Metformin can mitigate skeletal dysplasia caused by Pck2 deficiency. Int J Oral Sci 2022; 14:54. [PMCID: PMC9663691 DOI: 10.1038/s41368-022-00204-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Revised: 08/12/2022] [Accepted: 09/27/2022] [Indexed: 11/17/2022] Open
Abstract
As an important enzyme for gluconeogenesis, mitochondrial phosphoenolpyruvate carboxykinase (PCK2) has further complex functions beyond regulation of glucose metabolism. Here, we report that conditional knockout of Pck2 in osteoblasts results in a pathological phenotype manifested as craniofacial malformation, long bone loss, and marrow adipocyte accumulation. Ablation of Pck2 alters the metabolic pathways of developing bone, particularly fatty acid metabolism. However, metformin treatment can mitigate skeletal dysplasia of embryonic and postnatal heterozygous knockout mice, at least partly via the AMPK signaling pathway. Collectively, these data illustrate that PCK2 is pivotal for bone development and metabolic homeostasis, and suggest that regulation of metformin-mediated signaling could provide a novel and practical strategy for treating metabolic skeletal dysfunction.
Collapse
|
19
|
iPSC-neural crest derived cells embedded in 3D printable bio-ink promote cranial bone defect repair. Sci Rep 2022; 12:18701. [PMID: 36333414 PMCID: PMC9636385 DOI: 10.1038/s41598-022-22502-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 10/17/2022] [Indexed: 11/06/2022] Open
Abstract
Cranial bone loss presents a major clinical challenge and new regenerative approaches to address craniofacial reconstruction are in great demand. Induced pluripotent stem cell (iPSC) differentiation is a powerful tool to generate mesenchymal stromal cells (MSCs). Prior research demonstrated the potential of bone marrow-derived MSCs (BM-MSCs) and iPSC-derived mesenchymal progenitor cells via the neural crest (NCC-MPCs) or mesodermal lineages (iMSCs) to be promising cell source for bone regeneration. Overexpression of human recombinant bone morphogenetic protein (BMP)6 efficiently stimulates bone formation. The study aimed to evaluate the potential of iPSC-derived cells via neural crest or mesoderm overexpressing BMP6 and embedded in 3D printable bio-ink to generate viable bone graft alternatives for cranial reconstruction. Cell viability, osteogenic potential of cells, and bio-ink (Ink-Bone or GelXa) combinations were investigated in vitro using bioluminescent imaging. The osteogenic potential of bio-ink-cell constructs were evaluated in osteogenic media or nucleofected with BMP6 using qRT-PCR and in vitro μCT. For in vivo testing, two 2 mm circular defects were created in the frontal and parietal bones of NOD/SCID mice and treated with Ink-Bone, Ink-Bone + BM-MSC-BMP6, Ink-Bone + iMSC-BMP6, Ink-Bone + iNCC-MPC-BMP6, or left untreated. For follow-up, µCT was performed at weeks 0, 4, and 8 weeks. At the time of sacrifice (week 8), histological and immunofluorescent analyses were performed. Both bio-inks supported cell survival and promoted osteogenic differentiation of iNCC-MPCs and BM-MSCs in vitro. At 4 weeks, cell viability of both BM-MSCs and iNCC-MPCs were increased in Ink-Bone compared to GelXA. The combination of Ink-Bone with iNCC-MPC-BMP6 resulted in an increased bone volume in the frontal bone compared to the other groups at 4 weeks post-surgery. At 8 weeks, both iNCC-MPC-BMP6 and iMSC-MSC-BMP6 resulted in an increased bone volume and partial bone bridging between the implant and host bone compared to the other groups. The results of this study show the potential of NCC-MPC-incorporated bio-ink to regenerate frontal cranial defects. Therefore, this bio-ink-cell combination should be further investigated for its therapeutic potential in large animal models with larger cranial defects, allowing for 3D printing of the cell-incorporated material.
Collapse
|
20
|
Licini C, Notarstefano V, Marchi S, Cerqueni G, Ciapetti G, Vitale‐Brovarone C, Giorgini E, Mattioli‐Belmonte M. Altered type I collagen networking in osteoporotic human femoral head revealed by histomorphometric and Fourier transform infrared imaging correlated analyses. Biofactors 2022; 48:1089-1110. [PMID: 35661288 PMCID: PMC9796100 DOI: 10.1002/biof.1870] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 04/30/2022] [Indexed: 12/30/2022]
Abstract
Bone homeostasis is the equilibrium between organic and inorganic components of the extracellular matrix (ECM) and cells. Alteration of this balance has consequences on bone mass and architecture, resulting in conditions such as osteoporosis (OP). Given ECM protein mutual regulation and their effects on bone structure and mineralization, further insight into their expression is crucial to understanding bone biology under normal and pathological conditions. This study focused on Type I Collagen, which is mainly responsible for structural properties and mineralization of bone, and selected proteins implicated in matrix composition, mineral deposition, and cell-matrix interaction such as Decorin, Osteocalcin, Osteopontin, Bone Sialoprotein 2, Osteonectin and Transforming Growth Factor beta. We developed a novel multidisciplinary approach in order to assess bone matrix in healthy and OP conditions more comprehensively by exploiting the Fourier Transform Infrared Imaging (FTIRI) technique combined with histomorphometry, Sirius Red staining, immunohistochemistry, and Western Blotting. This innovatory procedure allowed for the analysis of superimposed tissue sections and revealed that the alterations in OP bone tissue architecture were associated with warped Type I Collagen structure and deposition but not with changes in the total protein amount. The detected changes in the expression and/or cooperative or antagonist role of Decorin, Osteocalcin, Osteopontin, and Bone Sialoprotein-2 indicate the deep impact of these NCPs on collagen features of OP bone. Overall, our strategy may represent a starting point for designing targeted clinical strategies aimed at bone mass preservation and sustain the FTIRI translational capability as upcoming support for traditional diagnostic methods.
Collapse
Affiliation(s)
- Caterina Licini
- Department of Clinical and Molecular Sciences (DISCLIMO)Università Politecnica delle MarcheAnconaItaly
- Department of Applied Science and TechnologyPolitecnico di TorinoTorinoItaly
| | - Valentina Notarstefano
- Department of Life and Environmental SciencesUniversità Politecnica delle MarcheAnconaItaly
| | - Saverio Marchi
- Department of Clinical and Molecular Sciences (DISCLIMO)Università Politecnica delle MarcheAnconaItaly
| | - Giorgia Cerqueni
- Department of Clinical and Molecular Sciences (DISCLIMO)Università Politecnica delle MarcheAnconaItaly
| | - Gabriela Ciapetti
- Laboratory of Nanobiotechnology (NaBi)IRCCS Istituto Ortopedico RizzoliBolognaItaly
| | | | - Elisabetta Giorgini
- Department of Life and Environmental SciencesUniversità Politecnica delle MarcheAnconaItaly
| | - Monica Mattioli‐Belmonte
- Department of Clinical and Molecular Sciences (DISCLIMO)Università Politecnica delle MarcheAnconaItaly
| |
Collapse
|
21
|
Zeng Y, Wang T, Liu Y, Luo T, Li Q, He Y, Fang M, He R. Wnt and Smad signaling pathways synergistically regulated the osteogenic differentiation of fibroblasts in ankylosing spondylitis. Tissue Cell 2022; 77:101852. [DOI: 10.1016/j.tice.2022.101852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 05/22/2022] [Accepted: 06/04/2022] [Indexed: 11/26/2022]
|
22
|
Abstract
Despite the significant progress made over the past decade with combination of molecular profiling data and the development of new clinical strategies, our understanding of metastasis remains elusive. Bone metastasis is a complex process and a major cause of mortality in breast and prostate cancer patients, for which there is no effective treatment to-date. The current review summarizes the routes taken by the metastatic cells and the interactions between them and the bone microenvironment. We emphasize the role of the specified niches and cues that promote cellular adhesion, colonization, prolonged dormancy, and reactivation. Understanding these mechanisms will provide better insights for future studies and treatment strategies for bone metastatic conditions.
Collapse
|
23
|
HDAC1 regulates inflammation and osteogenic differentiation of ankylosing spondylitis fibroblasts through the Wnt-Smad signaling pathway. J Orthop Surg Res 2022; 17:343. [PMID: 35794630 PMCID: PMC9258155 DOI: 10.1186/s13018-022-03224-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 06/18/2022] [Indexed: 11/10/2022] Open
Abstract
Ankylosing spondylitis (AS) is a refractory autoimmune disease, whose typical pathology is the development of inflammation to ossification and ankylosis. Histone deacetylase 1 (HDAC1) is considered to be a key factor involved in inflammatory gene transduction, but its role in AS remains unclear. The purpose of this study was to explore the role and possible mechanism of HDAC1 in AS based on the Wnt-Smad pathway. Fibroblasts were isolated from hip synovial tissues of AS patients, adeno-associated virus (AAV) was used to regulate the expression of HDAC1, DKK-1 and SIS3 was used to inhibit Wnt and Smad, respectively. The expressions of Wnt-Smad pathway-related proteins were analyzed by WB, and the TRP ion channel proteins were analyzed by immunofluorescence and WB. The proliferation of AS fibroblasts was detected by CCK-8, the expression of inflammatory cytokines was detected by ELISA, and the effects of HDAC1 on osteogenic differentiation of AS fibroblasts were investigated by alkaline phosphatase (ALP) activity, intracellular calcium concentration, mineralization and osteogenic proteins expressions. Results showed that HDAC1 significantly affected the protein expressions of the Wnt-Smad pathway in AS fibroblasts, and Wnt inhibitor DKK-1 and Smad3 inhibitor SIS3 could significantly reverse the effect of HDAC1 on the Wnt-Smad pathway. In addition, HDAC1 significantly activated the TRP ion channel and promoted the proliferation, inflammatory response and osteogenic differentiation of AS fibroblasts. DKK-1 or SIS3 treatment significantly inhibit the effect of HDAC-1 on AS fibroblasts, suggesting that the Wnt-Smad pathway is involved in the regulation of AS by HDAC1. In conclusion, HDAC1 promotes the proliferation, inflammatory response and osteogenic differentiation of AS fibroblasts through the Wnt-Smad pathway.
Collapse
|
24
|
Di Tinco R, Consolo U, Pisciotta A, Orlandi G, Bertani G, Nasi M, Bertacchini J, Carnevale G. Characterization of Dental Pulp Stem Cells Response to Bone Substitutes Biomaterials in Dentistry. Polymers (Basel) 2022; 14:polym14112223. [PMID: 35683895 PMCID: PMC9182880 DOI: 10.3390/polym14112223] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 05/26/2022] [Accepted: 05/28/2022] [Indexed: 02/05/2023] Open
Abstract
Bone substitute biomaterials (BSBs) represent a promising alternative to bone autografts, due to their biocompatibility, osteoconduction, slow resorption rates, and the ability to define and maintain volume for bone gain in dentistry. Many biomaterials are tailored to provide structural and biological support for bone regeneration, and allow the migration of bone-forming cells into the bone defect. Neural crest-derived stem cells isolated from human dental pulp (hDPSCs) represent a suitable stem cell source to study the biological effects of BSBs on osteoprogenitor cells involved in the physiological bone regenerative processes. This study aimed to evaluate how three different BSBs affect the stem cell properties, osteogenic differentiation, and inflammatory properties of hDPSCs. Our data highlight that BSBs do not alter cell proliferation and stemness markers expression, nor induce any inflammatory responses. Bone metabolism data show that hDPSCs exposed to the three BSBs distinctively secrete the factors supporting osteoblast activity and osteoclast activity. Our data indicate that (i) hDPSCs are a suitable stem cell source to study the effects of BSBs, and that (ii) the formulation of BSBs may condition the biological properties of stem cells, suggesting their versatile suitability to different dentistry applications.
Collapse
Affiliation(s)
- Rosanna Di Tinco
- Department of Surgery, Medicine, Dentistry and Morphological Sciences with Interest in Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, 41125 Modena, Italy; (R.D.T.); (U.C.); (A.P.); (G.O.); (G.B.); (M.N.); (J.B.)
| | - Ugo Consolo
- Department of Surgery, Medicine, Dentistry and Morphological Sciences with Interest in Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, 41125 Modena, Italy; (R.D.T.); (U.C.); (A.P.); (G.O.); (G.B.); (M.N.); (J.B.)
- Operative Unit of Dentistry and Maxillofacial Surgery, Department Integrated Activity-Specialist Surgeries, University-Hospital of Modena, 41125 Modena, Italy
| | - Alessandra Pisciotta
- Department of Surgery, Medicine, Dentistry and Morphological Sciences with Interest in Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, 41125 Modena, Italy; (R.D.T.); (U.C.); (A.P.); (G.O.); (G.B.); (M.N.); (J.B.)
| | - Giulia Orlandi
- Department of Surgery, Medicine, Dentistry and Morphological Sciences with Interest in Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, 41125 Modena, Italy; (R.D.T.); (U.C.); (A.P.); (G.O.); (G.B.); (M.N.); (J.B.)
| | - Giulia Bertani
- Department of Surgery, Medicine, Dentistry and Morphological Sciences with Interest in Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, 41125 Modena, Italy; (R.D.T.); (U.C.); (A.P.); (G.O.); (G.B.); (M.N.); (J.B.)
| | - Milena Nasi
- Department of Surgery, Medicine, Dentistry and Morphological Sciences with Interest in Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, 41125 Modena, Italy; (R.D.T.); (U.C.); (A.P.); (G.O.); (G.B.); (M.N.); (J.B.)
| | - Jessika Bertacchini
- Department of Surgery, Medicine, Dentistry and Morphological Sciences with Interest in Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, 41125 Modena, Italy; (R.D.T.); (U.C.); (A.P.); (G.O.); (G.B.); (M.N.); (J.B.)
- CNR-Institute of Molecular Genetics “Luigi Luca Cavalli-Sforza”, Unit of Bologna, 40136 Bologna, Italy
| | - Gianluca Carnevale
- Department of Surgery, Medicine, Dentistry and Morphological Sciences with Interest in Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, 41125 Modena, Italy; (R.D.T.); (U.C.); (A.P.); (G.O.); (G.B.); (M.N.); (J.B.)
- Correspondence: ; Tel.: +39-0594-224-852
| |
Collapse
|
25
|
Makishi S, Yamazaki T, Ohshima H. Osteopontin on the Dental Implant Surface Promotes Direct Osteogenesis in Osseointegration. Int J Mol Sci 2022; 23:ijms23031039. [PMID: 35162963 PMCID: PMC8835189 DOI: 10.3390/ijms23031039] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 01/08/2022] [Accepted: 01/15/2022] [Indexed: 02/01/2023] Open
Abstract
After dental implantation, osteopontin (OPN) is deposited on the hydroxyapatite (HA) blasted implant surface followed by direct osteogenesis, which is significantly disturbed in Opn-knockout (KO) mice. However, whether applying OPN on the implant surface promotes direct osteogenesis remains unclarified. This study analyzed the effects of various OPN modified protein/peptides coatings on the healing patterns of the bone-implant interface after immediately placed implantation in the maxilla of four-week-old Opn-KO and wild-type (WT) mice (n = 96). The decalcified samples were processed for immunohistochemistry for OPN and Ki67 and tartrate-resistant acid phosphatase histochemistry. In the WT mice, the proliferative activity in the HA binding peptide-OPN mimic peptide fusion coated group was significantly higher than that in the control group from day 3 to week 1, and the rates of OPN deposition and direct osteogenesis around the implant surface significantly increased in the recombinant-mouse-OPN (rOPN) group compared to the Gly-Arg-Gly-Asp-Ser peptide group in week 2. The rOPN group achieved the same rates of direct osteogenesis and osseointegration as those in the control group in a half period (week 2). None of the implant surfaces could rescue the direct osteogenesis in the healing process in the Opn-KO mice. These results suggest that the rOPN coated implant enhances direct osteogenesis during osseointegration following implantation.
Collapse
Affiliation(s)
- Sanako Makishi
- Division of Anatomy and Cell Biology of the Hard Tissue, Department of Tissue Regeneration and Reconstruction, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8514, Japan;
| | - Tomohiko Yamazaki
- Research Center for Functional Materials, National Institute for Materials Science, Tsukuba 305-0047, Japan;
| | - Hayato Ohshima
- Division of Anatomy and Cell Biology of the Hard Tissue, Department of Tissue Regeneration and Reconstruction, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8514, Japan;
- Correspondence: ; Tel.: +81-25-227-2812
| |
Collapse
|
26
|
Aslankoohi N, Lin S, Mequanint K. Bioactive fluorescent hybrid microparticles as a stand-alone osteogenic differentiation inducer. Mater Today Bio 2022; 13:100187. [PMID: 34977526 PMCID: PMC8683730 DOI: 10.1016/j.mtbio.2021.100187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 12/07/2021] [Accepted: 12/08/2021] [Indexed: 12/02/2022] Open
Abstract
Osteogenic differentiation of stem cells is one of the essential steps in bone regeneration. While supplementing exogenous factors using differentiation media is the established method to differentiate stem cells into osteoblasts on biomaterials, designing biomaterials that can act as a stand-alone differentiation inducer and promote bone regeneration is preferred for clinical translation. In this work, we report dexamethasone-loaded organic-inorganic hybrid microparticles synthesized from an intrinsically fluorescent poly (ester amide) and tertiary bioactive glass (PEA-BG) as a stand-alone osteogenic differentiation inducer. The mechanical properties data indicated that the compressive modulus of fluorescent hybrid microparticles could be modulated by its composition. The hybrid fluorescent microparticles supported the adhesion and proliferation of 10T1/2 cells in culture for up to seven days. Both pristine and dexamethasone-loaded PEA-BG microparticles were able to induce osteogenic differentiation of 10T1/2 cells in the absence of any media supplement, to a level even higher than standard osteogenic media, as evidenced by the expression of osteogenic markers on gene and protein levels and matrix mineralization. Taken together, the fluorescent PEA-BG hybrid microparticles have the potential to be used as a stand-alone biomaterial for osteogenic differentiation and bone regeneration.
Collapse
Affiliation(s)
- Neda Aslankoohi
- School of Biomedical Engineering, The University of Western Ontario, London, Ontario, N6A 5B9, Canada
| | - Shigang Lin
- Department of Chemical and Biochemical Engineering, The University of Western Ontario, London, Ontario, N6A 5B9, Canada
| | - Kibret Mequanint
- School of Biomedical Engineering, The University of Western Ontario, London, Ontario, N6A 5B9, Canada
- Department of Chemical and Biochemical Engineering, The University of Western Ontario, London, Ontario, N6A 5B9, Canada
| |
Collapse
|
27
|
Carvalho MS, Alves L, Bogalho I, Cabral JMS, da Silva CL. Impact of Donor Age on the Osteogenic Supportive Capacity of Mesenchymal Stromal Cell-Derived Extracellular Matrix. Front Cell Dev Biol 2021; 9:747521. [PMID: 34676216 PMCID: PMC8523799 DOI: 10.3389/fcell.2021.747521] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 09/10/2021] [Indexed: 12/27/2022] Open
Abstract
Mesenchymal stromal cells (MSC) have been proposed as an emerging cell-based therapeutic option for regenerative medicine applications as these cells can promote tissue and organ repair. In particular, MSC have been applied for the treatment of bone fractures. However, the healing capacity of these fractures is often compromised by patient's age. Therefore, considering the use of autologous MSC, we evaluated the impact of donor age on the osteogenic potential of bone marrow (BM)-derived MSC. MSC from older patients (60 and 80 years old) demonstrated impaired proliferative and osteogenic capacities compared to MSC isolated from younger patients (30 and 45 years old), suggesting that aging potentially changes the quantity and quality of MSC. Moreover, in this study, we investigated the capacity of the microenvironment [i.e., extracellular matrix (ECM)] to rescue the impaired proliferative and osteogenic potential of aged MSC. In this context, we aimed to understand if BM MSC features could be modulated by exposure to an ECM derived from cells obtained from young or old donors. When aged MSC were cultured on decellularized ECM derived from young MSC, their in vitro proliferative and osteogenic capacities were enhanced, which did not happen when cultured on old ECM. Our results suggest that the microenvironment, specifically the ECM, plays a crucial role in the quality (assessed in terms of osteogenic differentiation capacity) and quantity of MSC. Specifically, the aging of ECM is determinant of osteogenic differentiation of MSC. In fact, old MSC maintained on a young ECM produced higher amounts of extracellularly deposited calcium (9.10 ± 0.22 vs. 4.69 ± 1.41 μg.μl-1.10-7 cells for young ECM and old ECM, respectively) and up-regulated the expression of osteogenic gene markers such as Runx2 and OPN. Cell rejuvenation by exposure to a functional ECM might be a valuable clinical strategy to overcome the age-related decline in the osteogenic potential of MSC by recapitulating a younger microenvironment, attenuating the effects of aging on the stem cell niche. Overall, this study provides new insights on the osteogenic potential of MSC during aging and opens new possibilities for developing clinical strategies for elderly patients with limited bone formation capacity who currently lack effective treatments.
Collapse
Affiliation(s)
- Marta S. Carvalho
- Department of Bioengineering and IBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
| | - Laura Alves
- Department of Bioengineering and IBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
| | - Isabel Bogalho
- Department of Bioengineering and IBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
| | - Joaquim M. S. Cabral
- Department of Bioengineering and IBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
| | - Cláudia L. da Silva
- Department of Bioengineering and IBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
| |
Collapse
|
28
|
Um SH, Lee J, Song IS, Ok MR, Kim YC, Han HS, Rhee SH, Jeon H. Regulation of cell locomotion by nanosecond-laser-induced hydroxyapatite patterning. Bioact Mater 2021; 6:3608-3619. [PMID: 33869901 PMCID: PMC8022786 DOI: 10.1016/j.bioactmat.2021.03.025] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Revised: 03/09/2021] [Accepted: 03/09/2021] [Indexed: 02/08/2023] Open
Abstract
Hydroxyapatite, an essential mineral in human bones composed mainly of calcium and phosphorus, is widely used to coat bone graft and implant surfaces for enhanced biocompatibility and bone formation. For a strong implant-bone bond, the bone-forming cells must not only adhere to the implant surface but also move to the surface requiring bone formation. However, strong adhesion tends to inhibit cell migration on the surface of hydroxyapatite. Herein, a cell migration highway pattern that can promote cell migration was prepared using a nanosecond laser on hydroxyapatite coating. The developed surface promoted bone-forming cell movement compared with the unpatterned hydroxyapatite surface, and the cell adhesion and movement speed could be controlled by adjusting the pattern width. Live-cell microscopy, cell tracking, and serum protein analysis revealed the fundamental principle of this phenomenon. These findings are applicable to hydroxyapatite-coated biomaterials and can be implemented easily by laser patterning without complicated processes. The cell migration highway can promote and control cell movement while maintaining the existing advantages of hydroxyapatite coatings. Furthermore, it can be applied to the surface treatment of not only implant materials directly bonded to bone but also various implanted biomaterials implanted that require cell movement control.
Collapse
Affiliation(s)
- Seung-Hoon Um
- Center for Biomaterials, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
- Department of Dental Biomaterials Science, Dental Research Institute, School of Dentistry, Seoul National University, Seoul, 03080, Republic of Korea
| | - Jaehong Lee
- Center for Biomaterials, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - In-Seok Song
- Department of Oral and Maxillofacial Surgery, Korea University Anam Hospital, Seoul, 02841, Republic of Korea
| | - Myoung-Ryul Ok
- Center for Biomaterials, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Yu-Chan Kim
- Center for Biomaterials, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
- Division of Bio-Medical Science and Technology, KIST School, Korea University of Science and Technology, Seoul, 02792, Republic of Korea
| | - Hyung-Seop Han
- Center for Biomaterials, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Sang-Hoon Rhee
- Department of Dental Biomaterials Science, Dental Research Institute, School of Dentistry, Seoul National University, Seoul, 03080, Republic of Korea
| | - Hojeong Jeon
- Center for Biomaterials, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
- Division of Bio-Medical Science and Technology, KIST School, Korea University of Science and Technology, Seoul, 02792, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, Republic of Korea
| |
Collapse
|
29
|
Chaikiawkeaw D, Khorattanakulchai N, Nammultriputtar K, Rattanapisit K, Everts V, Kubera A, Phoolchareon W, Pavasant P. Osteopontin induces osteogenic differentiation by human periodontal ligament cells via calcium binding domain-activin receptor-like kinase (ALK-1) interaction. J Periodontol 2021; 93:e13-e23. [PMID: 34453745 DOI: 10.1002/jper.21-0184] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 07/28/2021] [Accepted: 08/09/2021] [Indexed: 01/03/2023]
Abstract
BACKGROUND Recently we have generated recombinant human osteopontin (rhOPN) using a plant platform (Nicotiana benthamiana) and demonstrated, when coated on culture plates, its osteogenic induction capacity of human periodontal ligament (PDL) cells. The aim of this study is to elucidate the molecular mechanism underlying the rhOPN-induced osteogenic differentiation of human PDL cells. METHODS Full length rhOPN (FL-OPN) and three constructs of OPN containing integrin binding domain (N142), calcium binding domain (C122) and mutated calcium-binding domain (C122δ) were generated from N. benthamiana. Human PDL cells were isolated from extracted third molars and cultured on FL-OPN, N142, C122, or C122δ-coated surfaces. Real-time PCR and Western blot analyses were used to determine mRNA and protein expression. In vitro calcification was determined by Alizarin red staining. A chemical inhibitor and RNAi silencing were used to elucidate signaling pathways. In silico analyses were performed to predict the protein-protein interaction. In vivo analysis was performed using a rat calvaria defect model. RESULTS Human PDL cells seeded on FL-OPN and C122-coated surfaces significantly increased both mRNA and protein expression of osterix (OSX) and enhanced in vitro calcification. Soluble FL-OPN as well as a surface coated with N142 did not affect OSX expression. Inhibition of activin receptor-like kinase (ALK-1) abolished the induction of osterix expression. In silico analysis suggested a possible interaction between the calcium binding domain (CaBD) of OPN and ALK-1 receptor. C122, but not C122δ coated surfaces, induced the expression of p-Smad-1 and this induction was inhibited by an ALK-1 inhibitor and RNAi against ALK-1. In vivo data showed that 3D porous scaffold containing C-122 enhanced new bone formation as compared to scaffold alone. CONCLUSION The results suggest that next to full length OPN, the CaBD of OPN, if coated to a surface, induces osteogenic differentiation via interaction with ALK-1 receptor.
Collapse
Affiliation(s)
- Daneeya Chaikiawkeaw
- Center of Excellence in Regenerative Dentistry, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Narach Khorattanakulchai
- Research Unit for Plant-Produced Pharmaceuticals, Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmaceutical Science, Chulalongkorn University, Bangkok, Thailand
| | - Ketsaraporn Nammultriputtar
- Center of Excellence in Regenerative Dentistry, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Kaewta Rattanapisit
- Research Unit for Plant-Produced Pharmaceuticals, Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmaceutical Science, Chulalongkorn University, Bangkok, Thailand
| | - Vincent Everts
- Center of Excellence in Regenerative Dentistry, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand.,Department of Oral Cell Biology, Academic Center of Dentistry Amsterdam, University of Amsterdam and Vrije Universiteit, Amsterdam, The Netherlands
| | - Anchanee Kubera
- Department of Genetics, Faculty of Science, Kasertsart University, Bangkok, Thailand
| | - Waranyoo Phoolchareon
- Research Unit for Plant-Produced Pharmaceuticals, Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmaceutical Science, Chulalongkorn University, Bangkok, Thailand
| | - Prasit Pavasant
- Center of Excellence in Regenerative Dentistry, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
30
|
Peng CH, Lin WY, Yeh KT, Chen IH, Wu WT, Lin MD. The molecular etiology and treatment of glucocorticoid-induced osteoporosis. Tzu Chi Med J 2021; 33:212-223. [PMID: 34386357 PMCID: PMC8323641 DOI: 10.4103/tcmj.tcmj_233_20] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 11/19/2020] [Accepted: 12/30/2020] [Indexed: 12/30/2022] Open
Abstract
Glucocorticoid-induced osteoporosis (GIOP) is the most common form of secondary osteoporosis, accounting for 20% of osteoporosis diagnoses. Using glucocorticoids for >6 months leads to osteoporosis in 50% of patients, resulting in an increased risk of fracture and death. Osteoblasts, osteocytes, and osteoclasts work together to maintain bone homeostasis. When bone formation and resorption are out of balance, abnormalities in bone structure or function may occur. Excess glucocorticoids disrupt the bone homeostasis by promoting osteoclast formation and prolonging osteoclasts' lifespan, leading to an increase in bone resorption. On the other hand, glucocorticoids inhibit osteoblasts' formation and facilitate apoptosis of osteoblasts and osteocytes, resulting in a reduction of bone formation. Several signaling pathways, signaling modulators, endocrines, and cytokines are involved in the molecular etiology of GIOP. Clinically, adults ≥40 years of age using glucocorticoids chronically with a high fracture risk are considered to have medical intervention. In addition to vitamin D and calcium tablet supplementations, the major therapeutic options approved for GIOP treatment include antiresorption drug bisphosphonates, parathyroid hormone N-terminal fragment teriparatide, and the monoclonal antibody denosumab. The selective estrogen receptor modulator can only be used under specific condition for postmenopausal women who have GIOP but fail to the regular GIOP treatment or have specific therapeutic contraindications. In this review, we focus on the molecular etiology of GIOP and the molecular pharmacology of the therapeutic drugs used for GIOP treatment.
Collapse
Affiliation(s)
- Cheng-Huan Peng
- Department of Orthopedics, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan.,Institute of Medical Sciences, Tzu Chi University, Hualien, Taiwan.,School of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Wen-Ying Lin
- Department of Orthopedics, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | - Kuang-Ting Yeh
- Department of Orthopedics, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan.,School of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Ing-Ho Chen
- Department of Orthopedics, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan.,School of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Wen-Tien Wu
- Department of Orthopedics, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan.,Institute of Medical Sciences, Tzu Chi University, Hualien, Taiwan.,School of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Ming-Der Lin
- Institute of Medical Sciences, Tzu Chi University, Hualien, Taiwan.,Department of Molecular Biology and Human Genetics, Tzu Chi University, Hualien, Taiwan
| |
Collapse
|
31
|
Huang Y, Liao L, Su H, Chen X, Jiang T, Liu J, Hou Q. Psoralen accelerates osteogenic differentiation of human bone marrow mesenchymal stem cells by activating the TGF-β/Smad3 pathway. Exp Ther Med 2021; 22:940. [PMID: 34306204 PMCID: PMC8281312 DOI: 10.3892/etm.2021.10372] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Accepted: 05/07/2021] [Indexed: 12/18/2022] Open
Abstract
Psoralen, one of the active ingredients in Psoralea corylifolia, has been previously reported to regulate the osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs). A previous study revealed that psoralen can regulate the expression levels of microRNA-488 and runt-related transcription factor 2 (Runx2) to promote the osteogenic differentiation of BMSCs. However, the underlying signalling pathway in this process remains to be fully elucidated. BMSCs have also been confirmed to play a key role in the occurrence and development of osteoporosis, and are expected to be potential seed cells in the treatment of osteoporosis. In order to explore the potential signalling pathways of psoralen acting on BMSCs, in the present study, human BMSCs (hBMSCs) were treated with different concentrations of psoralen (0.1, 1, 10 and 100 µmol/l) and the TGF-β receptor I (RI) inhibitor SB431542 (5 µmol/l) in vitro for 3, 7 or 14 days. Cell Counting Kit-8 and MTT assays were used to measure cell proliferation and cell viability of hBMSCs following psoralen administration. Alkaline phosphatase (ALP) activity and alizarin red S staining were used to assess the osteogenic differentiation ability of hBMSCs. Reverse transcription-quantitative PCR and western blotting were used to measure the expression of osteogenic differentiation-related genes [bone morphogenetic protein 4 (BMP4), osteopontin (OPN), Runx2 and Osterix] and proteins associated with the TGF-β/Smad3 pathway [TGF-β1, TGF-β RI, phosphorylated (p-)Smad and Smad3]. Psoralen was found to increase the proliferation and viability of hBMSCs. Although different concentrations of psoralen enhanced ALP activity and the calcified nodule content in hBMSCs, the enhancement effects were more potent at lower concentrations (0.1, 1 and 10 µmol/l). The expression of BMP4, OPN, Osterix, Runx2, TGF-β1, TGF-β RI and p-Smad3 was also promoted by psoralen at lower concentrations (0.1, 1 and 10 µmol/l). In addition, whilst SB431542 could inhibit calcium deposition and osteogenic differentiation-related gene expression in hBMSCs, psoralen effectively reversed the inhibitory effects of SB431542. In conclusion, psoralen accelerates the osteogenic differentiation of hBMSCs by activating the TGF-β/Smad3 pathway, which may be valuable for the future clinical treatment of osteoporosis.
Collapse
Affiliation(s)
- Yongquan Huang
- Department of Orthopaedics, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, P.R. China.,Graduate School, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510405, P.R. China
| | - Liu Liao
- Graduate School, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510405, P.R. China
| | - Haitao Su
- Department of Orthopaedics, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, P.R. China
| | - Xinlin Chen
- Department of Preventive Medicine and Health Statistics, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510405, P.R. China
| | - Tao Jiang
- Department of Orthopaedics, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, P.R. China
| | - Jun Liu
- Department of Orthopaedics, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, P.R. China
| | - Qiuke Hou
- Department of Gastroenterology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510405, P.R. China
| |
Collapse
|
32
|
Carvalho MS, Cabral JMS, da Silva CL, Vashishth D. Bone Matrix Non-Collagenous Proteins in Tissue Engineering: Creating New Bone by Mimicking the Extracellular Matrix. Polymers (Basel) 2021; 13:polym13071095. [PMID: 33808184 PMCID: PMC8036283 DOI: 10.3390/polym13071095] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 03/18/2021] [Accepted: 03/20/2021] [Indexed: 02/06/2023] Open
Abstract
Engineering biomaterials that mimic the extracellular matrix (ECM) of bone is of significant importance since most of the outstanding properties of the bone are due to matrix constitution. Bone ECM is composed of a mineral part comprising hydroxyapatite and of an organic part of primarily collagen with the rest consisting on non-collagenous proteins. Collagen has already been described as critical for bone tissue regeneration; however, little is known about the potential effect of non-collagenous proteins on osteogenic differentiation, even though these proteins were identified some decades ago. Aiming to engineer new bone tissue, peptide-incorporated biomimetic materials have been developed, presenting improved biomaterial performance. These promising results led to ongoing research focused on incorporating non-collagenous proteins from bone matrix to enhance the properties of the scaffolds namely in what concerns cell migration, proliferation, and differentiation, with the ultimate goal of designing novel strategies that mimic the native bone ECM for bone tissue engineering applications. Overall, this review will provide an overview of the several non-collagenous proteins present in bone ECM, their functionality and their recent applications in the bone tissue (including dental) engineering field.
Collapse
Affiliation(s)
- Marta S. Carvalho
- Center for Biotechnology and Interdisciplinary Studies, Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
- Department of Bioengineering and iBB—Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, 1049-001 Lisboa, Portugal; (J.M.S.C.); (C.L.d.S.)
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, 1049-001 Lisboa, Portugal
- Correspondence: (M.S.C.); (D.V.)
| | - Joaquim M. S. Cabral
- Department of Bioengineering and iBB—Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, 1049-001 Lisboa, Portugal; (J.M.S.C.); (C.L.d.S.)
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, 1049-001 Lisboa, Portugal
| | - Cláudia L. da Silva
- Department of Bioengineering and iBB—Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, 1049-001 Lisboa, Portugal; (J.M.S.C.); (C.L.d.S.)
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, 1049-001 Lisboa, Portugal
| | - Deepak Vashishth
- Center for Biotechnology and Interdisciplinary Studies, Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
- Correspondence: (M.S.C.); (D.V.)
| |
Collapse
|
33
|
Choi E, Bae S, Kim D, Yang GH, Lee K, You HJ, Kang HJ, Gwak SJ, An S, Jeon H. Characterization and intracellular mechanism of electrospun poly (ε-caprolactone) (PCL) fibers incorporated with bone-dECM powder as a potential membrane for guided bone regeneration. J IND ENG CHEM 2021. [DOI: 10.1016/j.jiec.2020.11.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
34
|
Živković JM, Stojanović ST, Vukelić-Nikolić MĐ, Radenković MB, Najdanović JG, Ćirić M, Najman SJ. Macrophages' contribution to ectopic osteogenesis in combination with blood clot and bone substitute: possibility for application in bone regeneration strategies. INTERNATIONAL ORTHOPAEDICS 2020; 45:1087-1095. [PMID: 33025084 DOI: 10.1007/s00264-020-04826-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 09/22/2020] [Indexed: 01/08/2023]
Abstract
PURPOSE Given the great potential of macrophages in the processes of tissue repair and regeneration, the aim of our study was to examine the contribution that macrophages will have in osteogenic process when combined and implanted with blood clot (BC) and mineral bone substitute (MBS) in mice subcutaneous implantation model. METHODS Three types of implants were constructed and implanted subcutaneously into BALB/c mice: (1) RMBM implants (made of resident tissue macrophages, BC and MBS), (2) BM implants (made of BC and MBS), and (3) M implants (made of MBS only) where the last two served as control implants. One, two, four and eight weeks after implantation implants were explanted, and histochemical, immunohistochemical, and histomorphometric analyses were performed. RESULTS Increased vascularization, particularly pronounced two and four weeks after implantation and pronounced tissue infiltration in eight week term in RMBM implants compared with both other types, likewise the presence of osteoblast-like cells, osteoid-like structures, and more prominent osteopontin and osteocalcin immunoexpression in RMBM implants indicated more pronounced osteogenic process within them. CONCLUSION Our results suggest that macrophages deserve to be considered as a cell component when constructing implants in bone regenerative medicine strategies to improve bone fracture healing process.
Collapse
Affiliation(s)
- Jelena M Živković
- Department of Biology and Human Genetics, Faculty of Medicine, University of Niš, Niš, 18108, Serbia. .,Department for Cell and Tissue Engineering, Scientific Research Center for Biomedicine, Faculty of Medicine, University of Niš, Niš, 18108, Serbia.
| | - Sanja T Stojanović
- Department of Biology and Human Genetics, Faculty of Medicine, University of Niš, Niš, 18108, Serbia.,Department for Cell and Tissue Engineering, Scientific Research Center for Biomedicine, Faculty of Medicine, University of Niš, Niš, 18108, Serbia
| | - Marija Đ Vukelić-Nikolić
- Department of Biology and Human Genetics, Faculty of Medicine, University of Niš, Niš, 18108, Serbia.,Department for Cell and Tissue Engineering, Scientific Research Center for Biomedicine, Faculty of Medicine, University of Niš, Niš, 18108, Serbia
| | - Milena B Radenković
- Department for Cell and Tissue Engineering, Scientific Research Center for Biomedicine, Faculty of Medicine, University of Niš, Niš, 18108, Serbia
| | - Jelena G Najdanović
- Department of Biology and Human Genetics, Faculty of Medicine, University of Niš, Niš, 18108, Serbia.,Department for Cell and Tissue Engineering, Scientific Research Center for Biomedicine, Faculty of Medicine, University of Niš, Niš, 18108, Serbia
| | - Milan Ćirić
- Institute of Physiology, Faculty of Medicine, University of Niš, Niš, 18108, Serbia
| | - Stevo J Najman
- Department of Biology and Human Genetics, Faculty of Medicine, University of Niš, Niš, 18108, Serbia.,Department for Cell and Tissue Engineering, Scientific Research Center for Biomedicine, Faculty of Medicine, University of Niš, Niš, 18108, Serbia
| |
Collapse
|
35
|
Lin X, Patil S, Gao YG, Qian A. The Bone Extracellular Matrix in Bone Formation and Regeneration. Front Pharmacol 2020; 11:757. [PMID: 32528290 PMCID: PMC7264100 DOI: 10.3389/fphar.2020.00757] [Citation(s) in RCA: 387] [Impact Index Per Article: 77.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 05/06/2020] [Indexed: 12/17/2022] Open
Abstract
Bone regeneration repairs bone tissue lost due to trauma, fractures, and tumors, or absent due to congenital disorders. The extracellular matrix (ECM) is an intricate dynamic bio-environment with precisely regulated mechanical and biochemical properties. In bone, ECMs are involved in regulating cell adhesion, proliferation, and responses to growth factors, differentiation, and ultimately, the functional characteristics of the mature bone. Bone ECM can induce the production of new bone by osteoblast-lineage cells, such as MSCs, osteoblasts, and osteocytes and the absorption of bone by osteoclasts. With the rapid development of bone regenerative medicine, the osteoinductive, osteoconductive, and osteogenic potential of ECM-based scaffolds has attracted increasing attention. ECM-based scaffolds for bone tissue engineering can be divided into two types, that is, ECM-modified biomaterial scaffold and decellularized ECM scaffold. Tissue engineering strategies that utilize the functional ECM are superior at guiding the formation of specific tissues at the implantation site. In this review, we provide an overview of the function of various types of bone ECMs in bone tissue and their regulation roles in the behaviors of osteoblast-lineage cells and osteoclasts. We also summarize the application of bone ECM in bone repair and regeneration. A better understanding of the role of bone ECM in guiding cellular behavior and tissue function is essential for its future applications in bone repair and regenerative medicine.
Collapse
Affiliation(s)
- Xiao Lin
- Laboratory for Bone Metabolism, Xi'an Key Laboratory of Special Medicine and Health Engineering, Key Laboratory for Space Biosciences and Biotechnology, Research Center for Special Medicine and Health Systems Engineering, NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Suryaji Patil
- Laboratory for Bone Metabolism, Xi'an Key Laboratory of Special Medicine and Health Engineering, Key Laboratory for Space Biosciences and Biotechnology, Research Center for Special Medicine and Health Systems Engineering, NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Yong-Guang Gao
- Laboratory for Bone Metabolism, Xi'an Key Laboratory of Special Medicine and Health Engineering, Key Laboratory for Space Biosciences and Biotechnology, Research Center for Special Medicine and Health Systems Engineering, NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Airong Qian
- Laboratory for Bone Metabolism, Xi'an Key Laboratory of Special Medicine and Health Engineering, Key Laboratory for Space Biosciences and Biotechnology, Research Center for Special Medicine and Health Systems Engineering, NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| |
Collapse
|
36
|
Carvalho MS, Silva JC, Hoff CM, Cabral JMS, Linhardt RJ, Silva CL, Vashishth D. Loss and rescue of osteocalcin and osteopontin modulate osteogenic and angiogenic features of mesenchymal stem/stromal cells. J Cell Physiol 2020; 235:7496-7515. [DOI: 10.1002/jcp.29653] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Accepted: 02/12/2020] [Indexed: 12/24/2022]
Affiliation(s)
- Marta S. Carvalho
- Department of Biomedical Engineering, Center for Biotechnology and Interdisciplinary StudiesRensselaer Polytechnic InstituteTroy New York
- Department of Bioengineering and iBB – Institute for Bioengineering and Biosciences, Instituto Superior TécnicoUniversidade de LisboaLisboa Portugal
| | - João C. Silva
- Department of Bioengineering and iBB – Institute for Bioengineering and Biosciences, Instituto Superior TécnicoUniversidade de LisboaLisboa Portugal
- Department of Chemistry and Chemical Biology, Biological Sciences and Chemical and Biological Engineering, Center for Biotechnology and Interdisciplinary StudiesRensselaer Polytechnic InstituteTroy New York
| | - Christopher M. Hoff
- Department of Earth and Environmental SciencesRensselaer Polytechnic InstituteTroy New York
| | - Joaquim M. S. Cabral
- Department of Bioengineering and iBB – Institute for Bioengineering and Biosciences, Instituto Superior TécnicoUniversidade de LisboaLisboa Portugal
| | - Robert J. Linhardt
- Department of Biomedical Engineering, Center for Biotechnology and Interdisciplinary StudiesRensselaer Polytechnic InstituteTroy New York
- Department of Chemistry and Chemical Biology, Biological Sciences and Chemical and Biological Engineering, Center for Biotechnology and Interdisciplinary StudiesRensselaer Polytechnic InstituteTroy New York
| | - Cláudia L. Silva
- Department of Bioengineering and iBB – Institute for Bioengineering and Biosciences, Instituto Superior TécnicoUniversidade de LisboaLisboa Portugal
| | - Deepak Vashishth
- Department of Biomedical Engineering, Center for Biotechnology and Interdisciplinary StudiesRensselaer Polytechnic InstituteTroy New York
| |
Collapse
|
37
|
The association between serum angiogenin and osteopontin levels and coronary collateral circulation in patients with chronic total occlusion. Anatol J Cardiol 2020; 22:77-84. [PMID: 31375653 PMCID: PMC6735440 DOI: 10.14744/anatoljcardiol.2019.88555] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
OBJECTIVE A well-developed coronary collateral circulation lowers both in-hospital and long-term morbidity and mortality limiting the infarct. Angiogenin (AGN) and osteopontin (OPN) are known to be potent inducers of angiogenesis. The aim of the present study was to investigate the relationship between serum ANG and OPN levels and collateral filling grade in subjects with stable coronary artery disease (SCAD). METHODS A total of 122 age- and gender-matched consecutive patients who were found to have total occlusion (n=70) and no significant stenosis in epicardial coronary arteries (n=52) who underwent coronary angiography due to SCAD between January 2015 and July 2017 were included in the study. AGN and OPN levels were measured using enzyme linked immunosorbent assay. Coronary collateral circulation was graded using Rentrop's classification of collateral filling. RESULTS A total of 52 patients (61.60±11.78 years, 61.5% male) without significant epicardial coronary artery stenosis and 70 patients (62.87±8.24 years, 65.7% male) with totally occluded coronary arteries were included in the study. Subjects with total occlusion had significantly higher levels of AGN [122.00 (79.00-623.00) pg/mL vs. 98.00 (18.00-160.00) pg/mL, p<0.001] and OPN [1863.50 (125.00-6500.00) pg/mL vs. 451.00 (112.00- 1850.00) pg/mL, p<0.001] than those without significant stenosis. In addition, AGN [127.00 (87.00-623.00) pg/mL vs. 110.00 (79.00-188.00) pg/mL, p=0.011] and OPN [2681.00 (126.00-6500.00) pg/mL vs. 649.00 (125.00-4255.00) pg/mL, p=0.001] levels were significantly higher in patients with better developed collaterals. Serum AGN and OPN levels were found to be significantly associated with coronary collateral development. CONCLUSION AGN and OPN are associated with better developed coronary collateral circulation and may have therapeutic implications for the promotion of coronary collateral development.
Collapse
|
38
|
In Vitro Evaluation of a Novel Osteo-Inductive Scaffold for Osteogenic Differentiation of Bone-Marrow Mesenchymal Stem Cells. J Craniofac Surg 2020; 31:577-582. [PMID: 31895856 DOI: 10.1097/scs.0000000000006133] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Demineralized bone matrices (DBMs) were demonstrated to be a promising candidate for bone regeneration by previous studies. However, the limited osteoinductivity of DBMs was insufficient for a better repairing of bone defect. Osteoblasts (OBs), the major cellular component of bone tissues, play an important role in the formation of new bone. The extracellular matrix (ECM) of OB is one of the main components of bone formation niche. OBJECTIVE To combine the DBMs with the ECM of OBs to construct a novel scaffold that could be used for bone reconstruction. METHODS In this study, OBs were cultured on the surface of DBMs for 10 days and removed by Triton X-100 and ammonium hydroxide to prepare the OBs-ECM-DBMs (OEDBMs). A series of material features such as residues of OBs and ECM, cytotoxity, and osteoinductive capability of OEDBMs were evaluated. RESULTS Low cell residues and low content of DNA were observed in OEDBMs. Compared with DBMs, OEDBMs possessed more bone tissues organic matrix proteins, such as osteocalcin, osteopontin, and collagen I. Rat bone marrow mesenchymal stem cells (rBMSCs) presented a good viability when cultured on both 2 materials. The significant upregulations of osteogenic genes and proteins of rBMSCs were observed in OEDBMs group compared with DBMs group. CONCLUSION Taken together, these findings suggested that the OB-secreted ECM may be qualified as an ideal modification method for enhancing the performance of engineered bone scaffold.
Collapse
|
39
|
Lamort AS, Giopanou I, Psallidas I, Stathopoulos GT. Osteopontin as a Link between Inflammation and Cancer: The Thorax in the Spotlight. Cells 2019; 8:cells8080815. [PMID: 31382483 PMCID: PMC6721491 DOI: 10.3390/cells8080815] [Citation(s) in RCA: 108] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 07/25/2019] [Accepted: 07/31/2019] [Indexed: 12/23/2022] Open
Abstract
The glycoprotein osteopontin (OPN) possesses multiple functions in health and disease. To this end, osteopontin has beneficial roles in wound healing, bone homeostasis, and extracellular matrix (ECM) function. On the contrary, osteopontin can be deleterious for the human body during disease. Indeed, osteopontin is a cardinal mediator of tumor-associated inflammation and facilitates metastasis. The purpose of this review is to highlight the importance of osteopontin in malignant processes, focusing on lung and pleural tumors as examples.
Collapse
Affiliation(s)
- Anne-Sophie Lamort
- Comprehensive Pneumology Center and Institute for Lung Biology and Disease, University Hospital, Ludwig-Maximilians University of Munich and Helmholtz Center Munich, Member of the German Center for Lung Research, Max-Lebsche-Platz 31, 81377 Munich, Bavaria, Germany.
| | - Ioanna Giopanou
- Laboratory for Molecular Respiratory Carcinogenesis, Department of Physiology, Faculty of Medicine, University of Patras, Biomedical Sciences Research Center, 1 Asklepiou Str., University Campus, 26504 Rio, Achaia, Greece
| | - Ioannis Psallidas
- Lungs for Living Research Centre, UCL Respiratory, University College London, London WC1E6BT, UK
| | - Georgios T Stathopoulos
- Comprehensive Pneumology Center and Institute for Lung Biology and Disease, University Hospital, Ludwig-Maximilians University of Munich and Helmholtz Center Munich, Member of the German Center for Lung Research, Max-Lebsche-Platz 31, 81377 Munich, Bavaria, Germany.
- Laboratory for Molecular Respiratory Carcinogenesis, Department of Physiology, Faculty of Medicine, University of Patras, Biomedical Sciences Research Center, 1 Asklepiou Str., University Campus, 26504 Rio, Achaia, Greece.
| |
Collapse
|