1
|
Doyle SE, Cazzola CN, Coleman CM. Design considerations when creating a high throughput screen-compatible in vitro model of osteogenesis. SLAS DISCOVERY : ADVANCING LIFE SCIENCES R & D 2024; 29:100184. [PMID: 39313131 DOI: 10.1016/j.slasd.2024.100184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 09/06/2024] [Accepted: 09/20/2024] [Indexed: 09/25/2024]
Abstract
Inducing osteogenic differentiation in vitro is useful for the identification and development of bone regeneration therapies as well as modelling bone disorders. To couple in vitro models with high throughput screening techniques retains the assay's relevance in research while increasing its therapeutic impact. Miniaturizing, automating and/or digitalizing in vitro assays will reduce the required quantity of cells, biologic stimulants, culture/output assay reagents, time and cost. This review highlights the design and workflow considerations for creating a high throughput screen-compatible model of osteogenesis, comparing and contrasting osteogenic cell type, assay fabrication and culture methodology, osteogenic induction approach and repurposing existing output techniques.
Collapse
Affiliation(s)
- Stephanie E Doyle
- Regenerative Medicine Institute, School of Medicine, College of Medicine, Nursing and Health Science, University of Galway, Galway City, County Galway H91 FD82, Ireland.
| | - Courtney N Cazzola
- Regenerative Medicine Institute, School of Medicine, College of Medicine, Nursing and Health Science, University of Galway, Galway City, County Galway H91 FD82, Ireland
| | - Cynthia M Coleman
- Regenerative Medicine Institute, School of Medicine, College of Medicine, Nursing and Health Science, University of Galway, Galway City, County Galway H91 FD82, Ireland
| |
Collapse
|
2
|
Patro BP, Jeyaraman N, Jayakumar T, Das G, Nallakumarasamy A, Jeyaraman M. Efficacy of Autologous Adult Live-Cultured Osteoblast (AALCO) Implantation in Avascular Necrosis of the Femoral Head: A Mid-Term Outcome Analysis. Indian J Orthop 2024; 58:1053-1063. [PMID: 39087043 PMCID: PMC11286887 DOI: 10.1007/s43465-024-01203-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Accepted: 06/03/2024] [Indexed: 08/02/2024]
Abstract
Introduction Avascular Necrosis (AVN) of the femoral head, a condition characterized by the interruption of blood supply leading to bone tissue death, presents significant therapeutic challenges. Recent advancements in orthobiologics, including the use of Autologous Adult Live-Cultured Osteoblasts (AALCO), combined with core decompression, offer a novel approach for managing AVN. This study assesses the efficacy of this treatment modality in improving functional outcomes and hindering disease progression. Materials and methods This retrospective observational study encompassed 30 patients treated between 2020 and 2023 for idiopathic AVN of the femoral head, grades I to III, who had not responded to conservative treatment. Patients were excluded based on specific criteria including age, secondary AVN causes, and certain health conditions. The treatment involved a two-stage surgical procedure under spinal anesthesia with OSSGROW® for AALCO generation. Post-operative care emphasized early mobilization, DVT prevention, and avoidance of NSAIDs. Outcome measures were evaluated using the Visual Analog Scale (VAS) for pain, modified Harris Hip Score, and annual MRI imaging for up to 36 months. Results Among 26 patients (41 hips) completing the study, statistically significant improvements in pain and hip functionality were documented, alongside positive radiological signs of osteogenesis in the majority of cases. However, four instances required advancement to total hip replacement due to disease progression. Conclusion The combination of core decompression and AALCO implantation shows promise as an effective treatment for AVN of the femoral head, with notable improvements in functional and radiological outcomes. This study supports the potential of orthobiologic approaches in AVN treatment, warranting further investigation through comprehensive randomized controlled trials. Graphical Abstract
Collapse
Affiliation(s)
- Bishnu Prasad Patro
- Department of Orthopaedics, All India Institute of Medical Sciences, Bhubaneswar, Odisha 751019 India
| | - Naveen Jeyaraman
- Department of Orthopaedics, ACS Medical College and Hospital, Dr MGR Educational and Research Institute, Chennai, Tamil Nadu 600077 India
| | - Tarun Jayakumar
- Department of Orthopaedics, KIMS-Sunshine Hospital, Hyderabad, Telangana 500003 India
| | - Gurudip Das
- Department of Orthopaedics, All India Institute of Medical Sciences, Bhubaneswar, Odisha 751019 India
| | - Arulkumar Nallakumarasamy
- Department of Orthopaedics, Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER), Karaikal, Puducherry, 609602 India
| | - Madhan Jeyaraman
- Department of Orthopaedics, ACS Medical College and Hospital, Dr MGR Educational and Research Institute, Chennai, Tamil Nadu 600077 India
| |
Collapse
|
3
|
Lau CS, Park SY, Ethiraj LP, Singh P, Raj G, Quek J, Prasadh S, Choo Y, Goh BT. Role of Adipose-Derived Mesenchymal Stem Cells in Bone Regeneration. Int J Mol Sci 2024; 25:6805. [PMID: 38928517 PMCID: PMC11204188 DOI: 10.3390/ijms25126805] [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: 04/15/2024] [Revised: 06/13/2024] [Accepted: 06/14/2024] [Indexed: 06/28/2024] Open
Abstract
Bone regeneration involves multiple factors such as tissue interactions, an inflammatory response, and vessel formation. In the event of diseases, old age, lifestyle, or trauma, bone regeneration can be impaired which could result in a prolonged healing duration or requiring an external intervention for repair. Currently, bone grafts hold the golden standard for bone regeneration. However, several limitations hinder its clinical applications, e.g., donor site morbidity, an insufficient tissue volume, and uncertain post-operative outcomes. Bone tissue engineering, involving stem cells seeded onto scaffolds, has thus been a promising treatment alternative for bone regeneration. Adipose-derived mesenchymal stem cells (AD-MSCs) are known to hold therapeutic value for the treatment of various clinical conditions and have displayed feasibility and significant effectiveness due to their ease of isolation, non-invasive, abundance in quantity, and osteogenic capacity. Notably, in vitro studies showed AD-MSCs holding a high proliferation capacity, multi-differentiation potential through the release of a variety of factors, and extracellular vesicles, allowing them to repair damaged tissues. In vivo and clinical studies showed AD-MSCs favoring better vascularization and the integration of the scaffolds, while the presence of scaffolds has enhanced the osteogenesis potential of AD-MSCs, thus yielding optimal bone formation outcomes. Effective bone regeneration requires the interplay of both AD-MSCs and scaffolds (material, pore size) to improve the osteogenic and vasculogenic capacity. This review presents the advances and applications of AD-MSCs for bone regeneration and bone tissue engineering, focusing on the in vitro, in vivo, and clinical studies involving AD-MSCs for bone tissue engineering.
Collapse
Affiliation(s)
- Chau Sang Lau
- National Dental Centre Singapore, National Dental Research Institute Singapore, Singapore 168938, Singapore; (C.S.L.); (S.Y.P.); (L.P.E.); (G.R.)
- Oral Health Academic Clinical Programme, Duke-NUS Medical School, Singapore 169857, Singapore
| | - So Yeon Park
- National Dental Centre Singapore, National Dental Research Institute Singapore, Singapore 168938, Singapore; (C.S.L.); (S.Y.P.); (L.P.E.); (G.R.)
| | - Lalith Prabha Ethiraj
- National Dental Centre Singapore, National Dental Research Institute Singapore, Singapore 168938, Singapore; (C.S.L.); (S.Y.P.); (L.P.E.); (G.R.)
- Oral Health Academic Clinical Programme, Duke-NUS Medical School, Singapore 169857, Singapore
| | - Priti Singh
- National Dental Centre Singapore, National Dental Research Institute Singapore, Singapore 168938, Singapore; (C.S.L.); (S.Y.P.); (L.P.E.); (G.R.)
| | - Grace Raj
- National Dental Centre Singapore, National Dental Research Institute Singapore, Singapore 168938, Singapore; (C.S.L.); (S.Y.P.); (L.P.E.); (G.R.)
| | - Jolene Quek
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore; (J.Q.); (Y.C.)
| | - Somasundaram Prasadh
- Center for Clean Energy Engineering, University of Connecticut, Storrs, CT 06269, USA;
| | - Yen Choo
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore; (J.Q.); (Y.C.)
| | - Bee Tin Goh
- National Dental Centre Singapore, National Dental Research Institute Singapore, Singapore 168938, Singapore; (C.S.L.); (S.Y.P.); (L.P.E.); (G.R.)
- Oral Health Academic Clinical Programme, Duke-NUS Medical School, Singapore 169857, Singapore
| |
Collapse
|
4
|
Macalester W, Boussahel A, Moreno-Tortolero RO, Shannon MR, West N, Hill D, Perriman A. A 3D In-vitro model of the human dentine interface shows long-range osteoinduction from the dentine surface. Int J Oral Sci 2024; 16:37. [PMID: 38734663 PMCID: PMC11088668 DOI: 10.1038/s41368-024-00298-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 03/18/2024] [Accepted: 03/18/2024] [Indexed: 05/13/2024] Open
Abstract
Emerging regenerative cell therapies for alveolar bone loss have begun to explore the use of cell laden hydrogels for minimally invasive surgery to treat small and spatially complex maxilla-oral defects. However, the oral cavity presents a unique and challenging environment for in vivo bone tissue engineering, exhibiting both hard and soft periodontal tissue as well as acting as key biocenosis for many distinct microbial communities that interact with both the external environment and internal body systems, which will impact on cell fate and subsequent treatment efficacy. Herein, we design and bioprint a facile 3D in vitro model of a human dentine interface to probe the effect of the dentine surface on human mesenchymal stem cells (hMSCs) encapsulated in a microporous hydrogel bioink. We demonstrate that the dentine substrate induces osteogenic differentiation of encapsulated hMSCs, and that both dentine and β-tricalcium phosphate substrates stimulate extracellular matrix production and maturation at the gel-media interface, which is distal to the gel-substrate interface. Our findings demonstrate the potential for long-range effects on stem cells by mineralized surfaces during bone tissue engineering and provide a framework for the rapid development of 3D dentine-bone interface models.
Collapse
Affiliation(s)
- William Macalester
- School of Cellular and Molecular Medicine, Biomedical Sciences Building, University of Bristol, University Walk, Bristol, United Kingdom
- Bristol Centre for Functional Nanomaterials, HH Wills Physics Laboratory, University of Bristol, Tyndall Avenue, Bristol, United Kingdom
| | - Asme Boussahel
- School of Cellular and Molecular Medicine, Biomedical Sciences Building, University of Bristol, University Walk, Bristol, United Kingdom.
| | - Rafael O Moreno-Tortolero
- Bristol Centre for Functional Nanomaterials, HH Wills Physics Laboratory, University of Bristol, Tyndall Avenue, Bristol, United Kingdom
- Centre for Protolife Research, School of Chemistry, University of Bristol, Cantocks Close, Bristol, United Kingdom
- Max Planck-Bristol Centre for Minimal Biology, School of Chemistry, University of Bristol, Bristol, United Kingdom
| | - Mark R Shannon
- School of Cellular and Molecular Medicine, Biomedical Sciences Building, University of Bristol, University Walk, Bristol, United Kingdom
| | - Nicola West
- Periodontology, Bristol Dental School, University of Bristol, Lower Maudlin Street, Bristol, United Kingdom
| | - Darryl Hill
- School of Cellular and Molecular Medicine, Biomedical Sciences Building, University of Bristol, University Walk, Bristol, United Kingdom
| | - Adam Perriman
- School of Cellular and Molecular Medicine, Biomedical Sciences Building, University of Bristol, University Walk, Bristol, United Kingdom.
| |
Collapse
|
5
|
Liu L, Hou S, Xu G, Gao J, Mu J, Gao M, He J, Su X, Yang Z, Liu Y, Chen T, Dong Z, Cheng L, Shi Z. Evaluation of osteogenic properties of a novel injectable bone-repair material containing strontium in vitro and in vivo. Front Bioeng Biotechnol 2024; 12:1390337. [PMID: 38707496 PMCID: PMC11069309 DOI: 10.3389/fbioe.2024.1390337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 04/11/2024] [Indexed: 05/07/2024] Open
Abstract
Objective: This study aims to develop and evaluate the biocompatibility and osteogenic potential of a novel injectable strontium-doped hydroxyapatite bone-repair material. Methods: The properties of strontium-doped hydroxyapatite/chitosan (Sr-HA/CS), hydroxyapatite/chitosan (HA/CS) and calcium phosphate/chitosan (CAP/CS) were assessed following their preparation via physical cross-linking and a one-step simplified method. Petri dishes containing Escherichia coli and Staphylococcus epidermidis were inoculated with the material for in vitro investigations. The material was also co-cultured with stem cells derived from human exfoliated deciduous teeth (SHEDs), to assess the morphology and proliferation capability of the SHEDs, Calcein-AM staining and the Cell Counting Kit-8 assay were employed. Osteogenic differentiation of SHEDs was determined using alkaline phosphatase (ALP) staining and Alizarin Red staining. For in vivo studies, Sr-HA/CS was implanted into the muscle pouch of mice and in a rat model of ovariectomy-induced femoral defects. Hematoxylin-eosin (HE) staining was performed to determine the extent of bone formation and defect healing. The formation of new bone was determined using Masson's trichrome staining. The osteogenic mechanism of the material was investigated using Tartrate-resistant acid phosphatase (TRAP) staining and immunohistochemical studies. Results: X-ray diffraction (XRD) and energy-dispersive spectroscopy (EDS) showed that strontium was successfully doped into HA. The Sr-HA/CS material can be uniformly squeezed using a syringe with a 13% swelling rate. Sr-HA/CS had a significant antibacterial effect against both E. coli and S. epidermidis (p < 0.05), with a stronger effect observed against E. coli. The Sr-HA/CS significantly improved cell proliferation and cell viability in vitro studies (p < 0.05). Compared to CAP/CS and CS, Sr-HA/CS generated a substantially greater new bone area during osteoinduction experiments (p < 0.05, p < 0.001). The Sr-HA/CS material demonstrated a significantly higher rate of bone repair in the bone defeat studies compared to the CAP/CS and CS materials (p < 0.01). The OCN-positive area and TRAP-positive cells in Sr-HA/CS were greater than those in control groups (p < 0.05). Conclusion: A novel injectable strontium-doped HA bone-repair material with good antibacterial properties, biocompatibility, and osteoinductivity was successfully prepared.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | - Lijia Cheng
- Clinical Medical College, Affiliated Hospital, School of Basic Medical Sciences of Chengdu University, Chengdu, China
| | - Zheng Shi
- Clinical Medical College, Affiliated Hospital, School of Basic Medical Sciences of Chengdu University, Chengdu, China
| |
Collapse
|
6
|
Chatzimentor I, Tsamesidis I, Ioannou ME, Pouroutzidou GK, Beketova A, Giourieva V, Papi R, Kontonasaki E. Study of Biological Behavior and Antimicrobial Properties of Cerium Oxide Nanoparticles. Pharmaceutics 2023; 15:2509. [PMID: 37896269 PMCID: PMC10610395 DOI: 10.3390/pharmaceutics15102509] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 10/11/2023] [Accepted: 10/20/2023] [Indexed: 10/29/2023] Open
Abstract
(1) Background: An element that has gained much attention in industrial and biomedical fields is Cerium (Ce). CeO2 nanoparticles have been proven to be promising regarding their different biomedical applications for the control of infection and inflammation. The aim of the present study was to investigate the biological properties and antimicrobial behavior of cerium oxide (CeO2) nanoparticles (NPs). (2) Methods: The investigation of the NPs' biocompatibility with human periodontal ligament cells (hPDLCs) was evaluated via the MTT assay. Measurement of alkaline phosphatase (ALP) levels and alizarine red staining (ARS) were used as markers in the investigation of CeO2 NPs' capacity to induce the osteogenic differentiation of hPDLCs. Induced inflammatory stress conditions were applied to hPDLCs with H2O2 to estimate the influence of CeO2 NPs on the viability of cells under these conditions, as well as to reveal any ROS scavenging properties. Total antioxidant capacity (TAC) of cell lysates with NPs was also investigated. Finally, the macro broth dilution method was the method of choice for checking the antibacterial capacity of CeO2 against the anaerobic pathogens Porphyromonas gingivalis and Prevotella intermedia. (3) Results: Cell viability assay indicated that hPDLCs increase their proliferation rate in a time-dependent manner in the presence of CeO2 NPs. ALP and ARS measurements showed that CeO2 NPs can promote the osteogenic differentiation of hPDLCs. In addition, the MTT assay and ROS determination demonstrated some interesting results concerning the viability of cells under oxidative stress conditions and, respectively, the capability of NPs to decrease free radical levels over the course of time. Antimicrobial toxicity was observed mainly against P. gingivalis. (4) Conclusions: CeO2 NPs could provide an excellent choice for use in clinical practices as they could prohibit bacterial proliferation and control inflammatory conditions.
Collapse
Affiliation(s)
- Iason Chatzimentor
- Department of Prosthodontics, Faculty of Health Sciences, School of Dentistry, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (I.C.); (I.T.); (M.-E.I.); (G.K.P.); (A.B.)
| | - Ioannis Tsamesidis
- Department of Prosthodontics, Faculty of Health Sciences, School of Dentistry, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (I.C.); (I.T.); (M.-E.I.); (G.K.P.); (A.B.)
| | - Maria-Eleni Ioannou
- Department of Prosthodontics, Faculty of Health Sciences, School of Dentistry, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (I.C.); (I.T.); (M.-E.I.); (G.K.P.); (A.B.)
| | - Georgia K. Pouroutzidou
- Department of Prosthodontics, Faculty of Health Sciences, School of Dentistry, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (I.C.); (I.T.); (M.-E.I.); (G.K.P.); (A.B.)
- Laboratory of Advanced Materials and Devices (AMDeLab), Faculty of Sciences, School of Physics, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Anastasia Beketova
- Department of Prosthodontics, Faculty of Health Sciences, School of Dentistry, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (I.C.); (I.T.); (M.-E.I.); (G.K.P.); (A.B.)
| | - Veronica Giourieva
- Laboratory of Biochemistry, Department of Chemistry, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (V.G.); (R.P.)
| | - Rigini Papi
- Laboratory of Biochemistry, Department of Chemistry, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (V.G.); (R.P.)
| | - Eleana Kontonasaki
- Department of Prosthodontics, Faculty of Health Sciences, School of Dentistry, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (I.C.); (I.T.); (M.-E.I.); (G.K.P.); (A.B.)
| |
Collapse
|
7
|
Wang C, Liu J, Min S, Liu Y, Liu B, Hu Y, Wang Z, Mao F, Wang C, Ma X, Wen P, Zheng Y, Tian Y. The effect of pore size on the mechanical properties, biodegradation and osteogenic effects of additively manufactured magnesium scaffolds after high temperature oxidation: An in vitro and in vivo study. Bioact Mater 2023; 28:537-548. [PMID: 37457041 PMCID: PMC10344631 DOI: 10.1016/j.bioactmat.2023.06.009] [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: 04/13/2023] [Revised: 05/30/2023] [Accepted: 06/16/2023] [Indexed: 07/18/2023] Open
Abstract
The effects of pore size in additively manufactured biodegradable porous magnesium on the mechanical properties and biodegradation of the scaffolds as well as new bone formation have rarely been reported. In this work, we found that high temperature oxidation improves the corrosion resistance of magnesium scaffold. And the effects of pore size on the mechanical characteristics and biodegradation of scaffolds, as well as new bone formation, were investigated using magnesium scaffolds with three different pore sizes, namely, 500, 800, and 1400 μm (P500, P800, and P1400). We discovered that the mechanical characteristics of the P500 group were much better than those of the other two groups. In vitro and in vivo investigations showed that WE43 magnesium alloy scaffolds supported the survival of mesenchymal stem cells and did not cause any local toxicity. Due to their larger specific surface area, the scaffolds in the P500 group released more magnesium ions within reasonable range and improved the osteogenic differentiation of bone mesenchymal stem cells compared with the other two scaffolds. In a rabbit femoral condyle defect model, the P500 group demonstrated unique performance in promoting new bone formation, indicating its great potential for use in bone defect regeneration therapy.
Collapse
Affiliation(s)
- Chaoxin Wang
- Department of Orthopedics, Peking University Third Hospital, Beijing, 100191, China
- Engineering Research Center of Bone and Joint Precision Medicine, Ministry of Education, Beijing, 100191, China
| | - Jinge Liu
- Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, China
| | - Shuyuan Min
- Department of Orthopedics, Peking University Third Hospital, Beijing, 100191, China
- Engineering Research Center of Bone and Joint Precision Medicine, Ministry of Education, Beijing, 100191, China
| | - Yu Liu
- Department of Orthopedics, Peking University Third Hospital, Beijing, 100191, China
- Engineering Research Center of Bone and Joint Precision Medicine, Ministry of Education, Beijing, 100191, China
| | - Bingchuan Liu
- Department of Orthopedics, Peking University Third Hospital, Beijing, 100191, China
- Engineering Research Center of Bone and Joint Precision Medicine, Ministry of Education, Beijing, 100191, China
| | - Yuanyu Hu
- Department of Orthopedics, Peking University Third Hospital, Beijing, 100191, China
- Engineering Research Center of Bone and Joint Precision Medicine, Ministry of Education, Beijing, 100191, China
| | - Zhengguang Wang
- Department of Orthopedics, Peking University Third Hospital, Beijing, 100191, China
- Engineering Research Center of Bone and Joint Precision Medicine, Ministry of Education, Beijing, 100191, China
| | - Fengbiao Mao
- Institute of Medicine Innovation and Research, Peking University Third Hospital, Beijing, 100191, China
| | - Caimei Wang
- Beijing AKEC Medical Co., Ltd., Beijing, 102200, China
| | - Xiaolin Ma
- Beijing AKEC Medical Co., Ltd., Beijing, 102200, China
| | - Peng Wen
- Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, China
| | - Yufeng Zheng
- School of Materials Science and Engineering, Peking University, Beijing, 100871, China
| | - Yun Tian
- Department of Orthopedics, Peking University Third Hospital, Beijing, 100191, China
- Engineering Research Center of Bone and Joint Precision Medicine, Ministry of Education, Beijing, 100191, China
| |
Collapse
|
8
|
Escobar LM, Bendahan Z, Garcia C, Castellanos JE. Relaxin treatment stimulates the differentiation of mesenchymal stem cells into osteoblasts. J Dent Sci 2023; 18:1786-1793. [PMID: 37799922 PMCID: PMC10548013 DOI: 10.1016/j.jds.2023.05.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Revised: 05/05/2023] [Indexed: 10/07/2023] Open
Abstract
Background/purpose Several studies have determined that relaxin stimulates differentiation and regulates the activity of mature osteoclasts, but little is known about its effect on the differentiation of mesenchymal cells towards the osteogenic lineage. Therefore, this study aimed to determine the effect of relaxin on the proliferation and differentiation of the osteoblastic lineage of mesenchymal cells derived from human dental pulp (hDPSC). Materials and methods In this in vitro study, hDPSC were characterized and treated with relaxin at different doses (10-80 ng/ml) and times (1-21 days). Morphology was assessed by microscopy, and proliferation was assessed using a resazurin assay. Osteoblastic differentiation was evaluated by Alizarin Red staining, alkaline phosphatase (ALP) labeling, and changes in the expression of the osteoblastic differentiation genes RUNX2 and BMP2. Results Relaxin treatment did not induce changes in the proliferation or viability of hDPSCs; however, larger cells and increased cytoplasmic prolongation were observed. Relaxin treatment (20 and 80 ng/ml) significantly increased calcified nodule formation on days 14 and 21. The cytochemical signals for ALP, RUNX2, and BMP2 gene expression were significantly (P < 0.05) increased by the relaxin treatment. Conclusion Relaxin treatment does not induce changes in hDPSC proliferation but induces morphological changes, increases ALP detection, calcified nodule formation, and increases expression of RUNX2 and BMP2, suggesting the induction of osteoblastic differentiation of hDPSC.
Collapse
Affiliation(s)
- Lina M. Escobar
- Grupo de Investigaciones Básicas y Aplicadas en Odontología, IBAPO Facultad de Odontología, Universidad Nacional de Colombia, Bogotá, Colombia
- Unidad de Manejo Integral de Malformaciones Craneofaciales UMIMC, Facultad de Odontología, Universidad El Bosque, Bogotá, Colombia
| | - Zita Bendahan
- Unidad de Manejo Integral de Malformaciones Craneofaciales UMIMC, Facultad de Odontología, Universidad El Bosque, Bogotá, Colombia
| | - Camilo Garcia
- Grupo de Investigaciones Básicas y Aplicadas en Odontología, IBAPO Facultad de Odontología, Universidad Nacional de Colombia, Bogotá, Colombia
- Grupo de Ortodoncia Actualizada en Investigación ORTOACTIV, Facultad de Odontología, Universidad Nacional de Colombia, Bogotá, Colombia
| | - Jaime E. Castellanos
- Grupo de Investigaciones Básicas y Aplicadas en Odontología, IBAPO Facultad de Odontología, Universidad Nacional de Colombia, Bogotá, Colombia
| |
Collapse
|
9
|
Nagai T, Sekimoto T, Kurogi S, Ohta T, Miyazaki S, Yamaguchi Y, Tajima T, Chosa E, Imasaka M, Yoshinobu K, Araki K, Araki M, Choijookhuu N, Sato K, Hishikawa Y, Funamoto T. Tmem161a regulates bone formation and bone strength through the P38 MAPK pathway. Sci Rep 2023; 13:14639. [PMID: 37670024 PMCID: PMC10480474 DOI: 10.1038/s41598-023-41837-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 08/31/2023] [Indexed: 09/07/2023] Open
Abstract
Bone remodeling is an extraordinarily complex process involving a variety of factors, such as genetic, metabolic, and environmental components. Although genetic factors play a particularly important role, many have not been identified. In this study, we investigated the role of transmembrane 161a (Tmem161a) in bone structure and function using wild-type (WT) and Tmem161a-depleted (Tmem161aGT/GT) mice. Mice femurs were examined by histological, morphological, and bone strength analyses. Osteoblast differentiation and mineral deposition were examined in Tmem161a-overexpressed, -knockdown and -knockout MC3T3-e1 cells. In WT mice, Tmem161a was expressed in osteoblasts of femurs; however, it was depleted in Tmem161aGT/GT mice. Cortical bone mineral density, thickness, and bone strength were significantly increased in Tmem161aGT/GT mice femurs. In MC3T3-e1 cells, decreased expression of alkaline phosphatase (ALP) and Osterix were found in Tmem161a overexpression, and these findings were reversed in Tmem161a-knockdown or -knockout cells. Microarray and western blot analyses revealed upregulation of the P38 MAPK pathway in Tmem161a-knockout cells, which referred as stress-activated protein kinases. ALP and flow cytometry analyses revealed that Tmem161a-knockout cells were resistant to oxidative stress. In summary, Tmem161a is an important regulator of P38 MAPK signaling, and depletion of Tmem161a induces thicker and stronger bones in mice.
Collapse
Affiliation(s)
- Takuya Nagai
- Division of Orthopaedic Surgery, Department of Medicine of Sensory and Motor Organs, Faculty of Medicine, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki, 889-1692, Japan
| | - Tomohisa Sekimoto
- Division of Orthopaedic Surgery, Department of Medicine of Sensory and Motor Organs, Faculty of Medicine, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki, 889-1692, Japan
| | - Syuji Kurogi
- Division of Orthopaedic Surgery, Department of Medicine of Sensory and Motor Organs, Faculty of Medicine, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki, 889-1692, Japan
| | - Tomomi Ohta
- Division of Orthopaedic Surgery, Department of Medicine of Sensory and Motor Organs, Faculty of Medicine, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki, 889-1692, Japan
| | - Shihoko Miyazaki
- Division of Orthopaedic Surgery, Department of Medicine of Sensory and Motor Organs, Faculty of Medicine, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki, 889-1692, Japan
| | - Yoichiro Yamaguchi
- Division of Orthopaedic Surgery, Department of Medicine of Sensory and Motor Organs, Faculty of Medicine, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki, 889-1692, Japan
| | - Takuya Tajima
- Division of Orthopaedic Surgery, Department of Medicine of Sensory and Motor Organs, Faculty of Medicine, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki, 889-1692, Japan
| | - Etsuo Chosa
- Division of Orthopaedic Surgery, Department of Medicine of Sensory and Motor Organs, Faculty of Medicine, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki, 889-1692, Japan
| | - Mai Imasaka
- Department of Genetics, Hyogo Medical University, Nishinomiya, Japan
| | - Kumiko Yoshinobu
- Institute of Resource Development and Analysis, Kumamoto University, Kumamoto, Japan
| | - Kimi Araki
- Institute of Resource Development and Analysis, Kumamoto University, Kumamoto, Japan
| | - Masatake Araki
- Institute of Resource Development and Analysis, Kumamoto University, Kumamoto, Japan
| | - Narantsog Choijookhuu
- Department of Anatomy, Histochemistry and Cell Biology, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
| | - Katsuaki Sato
- Division of Immunology Department of Infectious Disease, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
| | - Yoshitaka Hishikawa
- Department of Anatomy, Histochemistry and Cell Biology, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
| | - Taro Funamoto
- Division of Orthopaedic Surgery, Department of Medicine of Sensory and Motor Organs, Faculty of Medicine, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki, 889-1692, Japan.
| |
Collapse
|
10
|
Cell surface markers for mesenchymal stem cells related to the skeletal system: A scoping review. Heliyon 2023; 9:e13464. [PMID: 36865479 PMCID: PMC9970931 DOI: 10.1016/j.heliyon.2023.e13464] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 01/26/2023] [Accepted: 01/31/2023] [Indexed: 02/12/2023] Open
Abstract
Multipotent mesenchymal stromal cells (MSCs) have been described as bone marrow stromal cells, which can form cartilage, bone or hematopoietic supportive stroma. In 2006, the International Society for Cell Therapy (ISCT) established a set of minimal characteristics to define MSCs. According to their criteria, these cells must express CD73, CD90 and CD105 surface markers; however, it is now known they do not represent true stemness epitopes. The objective of the present work was to determine the surface markers for human MSCs associated with skeletal tissue reported in the literature (1994-2021). To this end, we performed a scoping review for hMSCs in axial and appendicular skeleton. Our findings determined the most widely used markers were CD105 (82.9%), CD90 (75.0%) and CD73 (52.0%) for studies performed in vitro as proposed by the ISCT, followed by CD44 (42.1%), CD166 (30.9%), CD29 (27.6%), STRO-1 (17.7%), CD146 (15.1%) and CD271 (7.9%) in bone marrow and cartilage. On the other hand, only 4% of the articles evaluated in situ cell surface markers. Even though most studies use the ISCT criteria, most publications in adult tissues don't evaluate the characteristics that establish a stem cell (self-renewal and differentiation), which will be necessary to distinguish between a stem cell and progenitor populations. Collectively, MSCs require further understanding of their characteristics if they are intended for clinical use.
Collapse
|
11
|
Ferreira‐Baptista C, Queirós A, Ferreira R, Fernandes MH, Gomes PS, Colaço B. Retinoic acid induces the osteogenic differentiation of cat adipose tissue-derived stromal cells from distinct anatomical sites. J Anat 2023; 242:277-288. [PMID: 36056547 PMCID: PMC9877480 DOI: 10.1111/joa.13758] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 07/07/2022] [Accepted: 08/16/2022] [Indexed: 02/01/2023] Open
Abstract
Mesenchymal stromal cells-based regenerative orthopedic therapies have been used in cats as a promising and innovative therapeutic approach to enhance the repair of bone defects. Adipose tissue-derived stromal cells (ADSCs) can be obtained from two main sites-subcutaneous and visceral-with established differences regarding structure, composition, cell content, and functionality. However, in cats, to the best of the authors' knowledge, no studies have been conducted to compare the functional activity of the ADSCs isolated from the two sites, and the impact of these differences on the induced osteogenic potential. Additionally, retinoic acid has been recently regarded as a new osteogenic inducer within cells of distinct species, with undisclosed functionality on cat-derived cell populations. Thus, the present study aimed to evaluate the functional activity of ADSCs isolated from the subcutaneous and visceral adipose sites (SCAT and VAT, respectively) of the cat, as well as the effects of two osteogenic-inducing conditions-the classic dexamethasone, β-glycerophosphate and ascorbic acid-supplemented media (Dex + β + AAM), and Retinoic Acid-supplemented media (RAM). The adipose tissue of subcutaneous and visceral origin was isolated, characterized, and ADSCs were isolated and grown in the presence of the two osteogenic-inducing conditions, and characterized in terms of proliferation, metabolic activity, morphology, and osteogenic activity. Our results demonstrated a distinct biological profile of the two adipose tissue sites regarding cell size, vascularization, and morphology. Further, osteogenic-induced ADSCs from both sites presented an increased expression of alkaline phosphatase activity (ALP) and cytochemical staining, as compared with control. Overall, RAM induced higher levels of ALP activity than Dex + β + AAM, supporting an increased osteogenic activation. Additionally, VAT was the tissue with the best osteogenic potential, showing higher levels of ALP expression, particularly with RAM. In conclusion, different characteristics were found between the two adipose tissue sites-SCAT and VAT, which probably reflect the differences found in the functionality of isolated ADSCs from both tissues. Furthermore, for cat, VAT shows a greater osteogenic-inductive capacity than SCAT, particularly with RAM, which can be of therapeutic relevance for regenerative medicine applications.
Collapse
Affiliation(s)
- Carla Ferreira‐Baptista
- Centre for the Research and Technology of Agro‐Environmental and Biological Sciences (CITAB)University of Trás‐os‐Montes e Alto Douro (UTAD)Vila RealPortugal
- BoneLab ‐ Laboratory for Bone Metabolism and Regeneration, Faculty of Dental MedicineUniversity of PortoPortoPortugal
- REQUIMTE/LAQVUniversity of PortoPortoPortugal
- REQUIMTE/LAQVDepartment of Chemistry University of AveiroAveiroPortugal
| | | | - Rita Ferreira
- REQUIMTE/LAQVDepartment of Chemistry University of AveiroAveiroPortugal
| | - Maria Helena Fernandes
- BoneLab ‐ Laboratory for Bone Metabolism and Regeneration, Faculty of Dental MedicineUniversity of PortoPortoPortugal
- REQUIMTE/LAQVUniversity of PortoPortoPortugal
| | - Pedro Sousa Gomes
- BoneLab ‐ Laboratory for Bone Metabolism and Regeneration, Faculty of Dental MedicineUniversity of PortoPortoPortugal
- REQUIMTE/LAQVUniversity of PortoPortoPortugal
| | - Bruno Colaço
- Centre for the Research and Technology of Agro‐Environmental and Biological Sciences (CITAB)University of Trás‐os‐Montes e Alto Douro (UTAD)Vila RealPortugal
- REQUIMTE/LAQVUniversity of PortoPortoPortugal
- CECAV—Animal and Veterinary Research Centre UTADUniversity of Trás‐os‐Montes and Alto DouroVila RealPortugal
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS)Vila RealPortugal
| |
Collapse
|
12
|
Embedding Silk Fibroin-Alginate Hydrogel in a 3D-Printed Porous Poly(Lactic Acid) Bone Tissue Scaffold Augments Stem Cell Function. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2023. [DOI: 10.1007/s40883-022-00286-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
|
13
|
Torrents S, Grau-Vorster M, Vives J. Potency Assays: The 'Bugaboo' of Stem Cell Therapy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1420:29-38. [PMID: 37258782 DOI: 10.1007/978-3-031-30040-0_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
Substantially manipulated cell-based products for human use are considered medicines and therefore regulatory authorities require extensive characterisation in terms of identity, purity and potency. The latter critical quality attribute is probably the most challenging to identify and measure, requiring provision that potency assays should reflect the intended mechanism of action and demonstrate the drugs' biological effect. However, in most cases, the mechanisms involved are not fully understood, making the definition and validation of suitable potency tests difficult, a 'bugaboo' quest to be feared. Although it is evident that much work is still needed in the scientific arena, the present chapter focuses on strategies currently used by developers of cell- and gene-based therapies to demonstrate potency of innovative medicines, the regulatory framework and need for standardisation seeking to demystify critical factors to consider when designing a potency assay.
Collapse
Affiliation(s)
- Sílvia Torrents
- Banc de Sang i Teixits, Edifici Dr. Frederic Duran i Jordà, Barcelona, Spain
- Transfusion Medicine group, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Marta Grau-Vorster
- Banc de Sang i Teixits, Edifici Dr. Frederic Duran i Jordà, Barcelona, Spain
- Transfusion Medicine group, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Joaquim Vives
- Banc de Sang i Teixits, Edifici Dr. Frederic Duran i Jordà, Barcelona, Spain.
- Musculoskeletal Tissue Engineering Group, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain.
- Departament de Medicina, Universitat Autònoma de Barcelona, Barcelona, Spain.
| |
Collapse
|
14
|
Osseointegration Properties of Titanium Implants Treated by Nonthermal Atmospheric-Pressure Nitrogen Plasma. Int J Mol Sci 2022; 23:ijms232315420. [PMID: 36499747 PMCID: PMC9740438 DOI: 10.3390/ijms232315420] [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: 11/18/2022] [Revised: 12/01/2022] [Accepted: 12/03/2022] [Indexed: 12/12/2022] Open
Abstract
Pure titanium is used in dental implants owing to its excellent biocompatibility and physical properties. However, the aging of the material during storage is detrimental to the long-term stability of the implant after implantation. Therefore, in this study, we attempted to improve the surface properties and circumvent the negative effects of material aging on titanium implants by using a portable handheld nonthermal plasma device capable of piezoelectric direct discharge to treat pure titanium discs with nitrogen gas. We evaluated the osteogenic properties of the treated samples by surface morphology and elemental analyses, as well as in vitro and in vivo experiments. The results showed that nonthermal atmospheric-pressure nitrogen plasma can improve the hydrophilicity of pure titanium without damaging its surface morphology while introducing nitrogen-containing functional groups, thereby promoting cell attachment, proliferation, and osseointegration to some extent. Therefore, nitrogen plasma treatment may be a promising method for the rapid surface treatment of titanium implants.
Collapse
|
15
|
Bispo DSC, Jesus CSH, Romek K, Marques IMC, Oliveira MB, Mano JF, Gil AM. An Intracellular Metabolic Signature as a Potential Donor-Independent Marker of the Osteogenic Differentiation of Adipose Tissue Mesenchymal Stem Cells. Cells 2022; 11:cells11233745. [PMID: 36497004 PMCID: PMC9739047 DOI: 10.3390/cells11233745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 11/17/2022] [Accepted: 11/19/2022] [Indexed: 11/25/2022] Open
Abstract
This paper describes an untargeted NMR metabolomics study to identify potential intracellular donor-dependent and donor-independent metabolic markers of proliferation and osteogenic differentiation of human adipose mesenchymal stem cells (hAMSCs). The hAMSCs of two donors with distinct proliferating/osteogenic characteristics were fully characterized regarding their polar endometabolome during proliferation and osteogenesis. An 18-metabolites signature (including changes in alanine, aspartate, proline, tyrosine, ATP, and ADP, among others) was suggested to be potentially descriptive of cell proliferation, independently of the donor. In addition, a set of 11 metabolites was proposed to compose a possible donor-independent signature of osteogenesis, mostly involving changes in taurine, glutathione, methylguanidine, adenosine, inosine, uridine, and creatine/phosphocreatine, choline/phosphocholine and ethanolamine/phosphocholine ratios. The proposed signatures were validated for a third donor, although they require further validation in a larger donor cohort. We believe that this proof of concept paves the way to exploit metabolic markers to monitor (and potentially predict) cell proliferation and the osteogenic ability of different donors.
Collapse
|
16
|
Farmani AR, Nekoofar MH, Ebrahimi-Barough S, Azami M, Najafipour S, Moradpanah S, Ai J. Preparation and In Vitro Osteogenic Evaluation of Biomimetic Hybrid Nanocomposite Scaffolds Based on Gelatin/Plasma Rich in Growth Factors (PRGF) and Lithium-Doped 45s5 Bioactive Glass Nanoparticles. JOURNAL OF POLYMERS AND THE ENVIRONMENT 2022; 31:870-885. [PMID: 36373108 PMCID: PMC9638231 DOI: 10.1007/s10924-022-02615-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Accepted: 09/26/2022] [Indexed: 06/16/2023]
Abstract
Bone tissue engineering is an emerging technique for repairing large bone lesions. Biomimetic techniques expand the use of organic-inorganic spongy-like nanocomposite scaffolds and platelet concentrates. In this study, a biomimetic nanocomposite scaffold was prepared using lithium-doped bioactive-glass nanoparticles and gelatin/PRGF. First, sol-gel method was used to prepare bioactive-glass nanoparticles that contain 0, 1, 3, and 5%wt lithium. The lithium content was then optimized based on antibacterial and MTT testing. By freeze-drying, hybrid scaffolds comprising 5, 10, and 20% bioglass were made. On the scaffolds, human endometrial stem cells (hEnSCs) were cultured for adhesion (SEM), survival, and osteogenic differentiation. Alkaline phosphatase activity and osteopontin, osteocalcin, and Runx2 gene expression were measured. The effect of bioactive-glass nanoparticles and PRGF on nanocomposites' mechanical characteristics and glass-transition temperature (T g) was also studied. An optimal lithium content in bioactive glass structure was found to be 3% wt. Nanoparticle SEM examination indicated grain deformation due to different sizes of lithium and sodium ions. Results showed up to 10% wt bioactive-glass and PRGF increased survival and cell adhesion. Also, Hybrid scaffolds revealed higher ALP-activity and OP, OC, and Runx2 gene expression. Furthermore, bioactive-glass has mainly increased ALP-activity and Runx2 expression, whereas PRGF increases the expression of OP and OC genes. Bioactive-glass increases scaffold modulus and T g continuously. Hence, the presence of both bioactive-glass and nanocomposite scaffold improves the expression of osteogenic differentiation biomarkers. Subsequently, it seems that hybrid scaffolds based on biopolymers, Li-doped bioactive-glass, and platelet extracts can be a good strategy for bone repair.
Collapse
Affiliation(s)
- Ahmad Reza Farmani
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Fasa University of Medical Sciences, Fasa, Iran
- Students’ Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Hossein Nekoofar
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Department of Endodontics, School of Dentistry, Tehran University of Medical Sciences, Tehran, Iran
- Department of Endodontics, School of Dentistry, Bahçeşehir University, Istanbul, Turkey
| | - Somayeh Ebrahimi-Barough
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahmoud Azami
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Sohrab Najafipour
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Fasa University of Medical Sciences, Fasa, Iran
- Department of Microbiology, School of Medicine, Fasa University of Medical Sciences, Fasa, Iran
| | - Somayeh Moradpanah
- Department of Obstetrics and Gynecology, Ziaeian Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Jafar Ai
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
17
|
Schott NG, Vu H, Stegemann JP. Multimodular vascularized bone construct comprised of vasculogenic and osteogenic microtissues. Biotechnol Bioeng 2022; 119:3284-3296. [PMID: 35922969 PMCID: PMC9547967 DOI: 10.1002/bit.28201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 07/17/2022] [Accepted: 07/30/2022] [Indexed: 01/05/2023]
Abstract
Bioengineered bone designed to heal large defects requires concomitant development of osseous and vascular tissue to ensure engraftment and survival. Adult human mesenchymal stromal cells (MSC) are promising in this application because they have demonstrated both osteogenic and vasculogenic potential. This study employed a modular approach in which cells were encapsulated in biomaterial carriers (microtissues) designed to support tissue-specific function. Osteogenic microtissues consisting of MSC embedded in a collagen-chitosan matrix; vasculogenic (VAS) microtissues consisted of endothelial cells and MSC in a fibrin matrix. Microtissues were precultured under differentiation conditions to induce appropriate MSC lineage commitment, and were then combined in a surrounding fibrin hydrogel to create a multimodular construct. Results demonstrated the ability of microtissues to support lineage commitment, and that preculture primes the microtissues for the desired function. Combination of osteogenic and vasculogenic microtissues into multimodular constructs demonstrated that osteogenic priming resulted in sustained osteogenic activity even when cultured in vasculogenic medium, and that vasculogenic priming induced a pericyte-like phenotype that resulted in development of a primitive vessel network in the constructs. The modular approach allows microtissues to be separately precultured to harness the dual differentiation potential of MSC to support both bone and blood vessel formation in a unified construct.
Collapse
Affiliation(s)
- Nicholas G. Schott
- Department of Biomedical EngineeringUniversity of MichiganAnn ArborMichiganUSA
| | - Huy Vu
- Department of Biomedical EngineeringUniversity of MichiganAnn ArborMichiganUSA
| | - Jan P. Stegemann
- Department of Biomedical EngineeringUniversity of MichiganAnn ArborMichiganUSA
| |
Collapse
|
18
|
Sun Q, Yu L, Zhang Z, Qian C, Fang H, Wang J, Wu P, Zhu X, Zhang J, Zhong L, He R. A novel gelatin/carboxymethyl chitosan/nano-hydroxyapatite/β-tricalcium phosphate biomimetic nanocomposite scaffold for bone tissue engineering applications. Front Chem 2022; 10:958420. [PMID: 36157039 PMCID: PMC9493496 DOI: 10.3389/fchem.2022.958420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 07/28/2022] [Indexed: 11/23/2022] Open
Abstract
Hydroxyapatite (HA) and tricalcium phosphate (TCP) constitute 60% of the content of the bone, and their combination has a better effect on bone tissue engineering than either single element. This study demonstrates a new degradable gelatin/carboxymethyl chitosan (CMC) bone scaffold loaded with both nano-HA and β-TCP (hereinafter referred to as HCP), and freeze drying combined with stir foaming was used to obtain highly connected macropores. Only a few studies have used these components to synthesize a four-component osteogenic scaffold. The aim of this study was to comprehensively assess the biocompatibility and osteoinductivity of the nanocomposites. Three HCP/CMC/gelatin scaffolds were made with different HCP contents: group A (10 wt% HCP), group B (30 wt% HCP), and group C (50 wt% HCP) (the ratio of nano-HA and β-TCP was fixed at 3:2). The scaffolds were macroporous with a high porosity and pore connectivity, as observed by morphological analysis by scanning electron microscopy. Additionally, the pore size of groups A and B was more homogeneous than that of group C. There were no significant differences in physicochemical characterization among the three groups. The Fourier-transform infrared (FTIR) spectroscopy test indicated that the scaffold contained active groups, such as hydroxyl, amino, or peptide bonds, corresponding to gelatin and CMC. The XRD results showed that the phase structures of HA and β-TCP did not change in the nanocomposite. The scaffolds had biodegradation potential and an appreciable swelling ratio, as demonstrated with the in vitro test. The scaffolds were cultured in vitro with MC3T3-E1 cells, showing that osteoinduction and osteoconduction increased with the HCP content. None of the scaffolds showed cytotoxicity. However, cell adhesion and growth in group B were better than those in group A and group C. Therefore, freeze drying combined with a stir foaming method may have a solid component limit. This study demonstrates a novel four-component scaffold via a simple manufacturing process. Group B (30% HCP) had the best characteristics for bone scaffold materials.
Collapse
Affiliation(s)
- Qiushuo Sun
- School of Stomatology, Hangzhou Normal University, Hangzhou, China
| | - Lu Yu
- School of Stomatology, Hangzhou Normal University, Hangzhou, China
| | - Zhuocheng Zhang
- School of Stomatology, Hangzhou Normal University, Hangzhou, China
| | - Cheng Qian
- School of Stomatology, Hangzhou Normal University, Hangzhou, China
| | - Hongzhe Fang
- School of Stomatology, Hangzhou Normal University, Hangzhou, China
| | - Jintao Wang
- Center of Stomatology, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, China
| | - Peipei Wu
- Center of Stomatology, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, China
| | - Xiaojing Zhu
- Institute of Life Sciences, College of Life and Environmental Sciences, Key Laboratory of Mammalian Organogenesis and Regeneration, Hangzhou Normal University, Hangzhou, China
| | - Jian Zhang
- College of Materials, Chemistry and Chemical Engineering, Hangzhou Normal University, Hangzhou, China
| | - Liangjun Zhong
- School of Stomatology, Hangzhou Normal University, Hangzhou, China
- Center of Stomatology, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, China
| | - Rui He
- School of Stomatology, Hangzhou Normal University, Hangzhou, China
- Center of Stomatology, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, China
- *Correspondence: Rui He,
| |
Collapse
|
19
|
Salam N, Gibson IR. Lithium ion doped carbonated hydroxyapatite compositions: Synthesis, physicochemical characterisation and effect on osteogenic response in vitro. BIOMATERIALS ADVANCES 2022; 140:213068. [PMID: 35939955 DOI: 10.1016/j.bioadv.2022.213068] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 07/08/2022] [Accepted: 08/01/2022] [Indexed: 06/15/2023]
Abstract
Hydroxyapatite is a commonly researched biomaterial for bone regeneration applications. To augment performance, hydroxyapatite can be substituted with functional ions to promote repair. Here, co-substituted lithium ion (Li+) and carbonate ion hydroxyapatite compositions were synthesised by an aqueous precipitation method. The co-substitution of Li+ and CO32- is a novel approach that accounts for charge balance, which has been ignored in the synthesis of Li doped calcium phosphates to date. Three compositions were synthesised: Li+-free (Li 0), low Li+ (Li 0.25), and high Li+ (Li 1). Synthesised samples were sintered as microporous discs (70-75 % theoretical sintered density) prior to being ground and fractionated to produce granules and powders, which were then characterised and evaluated in vitro. Physical and chemical characterisation demonstrated that lithium incorporation in Li 0.25 and Li 1 samples approached design levels (0.25 and 1 mol%), containing 0.253 and 0.881 mol% Li+ ions, respectively. The maximum CO32- ion content was observed in the Li 1 sample, with ~8 wt% CO3, with the carbonate ions located on both phosphate and hydroxyl sites in the crystal structure. Measurement of dissolution products following incubation experiments indicated a Li+ burst release profile in DMEM, with incubation of 30 mg/ml sample resulting in a Li+ ion concentration of approximately 140 mM after 24 h. For all compositions evaluated, sintered discs allowed for favourable attachment and proliferation of C2C12 cells, human osteoblast (hOB) cells, and human mesenchymal stem cells (hMSCs). An increase in alkaline phosphatase (ALP) activity with Li+ doping was demonstrated in C2C12 cells and hMSCs seeded onto sintered discs, whilst the inverse was observed in hOB cells. Furthermore, an increase in ALP activity was observed in C2C12 cells and hMSCs in response to dissolution products from Li 1 samples which related to Li+ release. Complementary experiments to further investigate the findings from hOB cells confirmed an osteogenic role of the surface topography of the discs. This research has shown successful synthesis of Li+ doped carbonated hydroxyapatite which demonstrated cytocompatibility and enhanced osteogenesis in vitro, compared to Li+-free controls.
Collapse
Affiliation(s)
- Nasseem Salam
- Aberdeen Centre for Arthritis and Musculoskeletal Health, Institute of Medical Sciences, University of Aberdeen, Aberdeen AB25 2ZD, UK
| | - Iain R Gibson
- Aberdeen Centre for Arthritis and Musculoskeletal Health, Institute of Medical Sciences, University of Aberdeen, Aberdeen AB25 2ZD, UK; Department of Chemistry, University of Aberdeen, Meston Walk, Aberdeen AB24 3UE, UK.
| |
Collapse
|
20
|
Liu M, Jin F, Zhang S, Li S, Zhu D, Cui Y, Cai M, Liu X, Zhang Y, Sun Y, Liu C, Wang X. Activation of farnesoid X receptor signaling by geniposidic acid promotes osteogenesis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 103:154258. [PMID: 35716540 DOI: 10.1016/j.phymed.2022.154258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 05/26/2022] [Accepted: 06/07/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND New targets and strategies are urgently needed for the identification and development of anabolic drugs for osteoporosis. Farnesoid X receptor (FXR) is a promising novel therapeutic target for bone metabolism diseases. Although used clinically, FXR agonists have obvious side effects; therefore, the development of new FXR agonists for the treatment of osteoporosis would be welcomed. Geniposidic acid (GPA) is a bioactive compound extracted from Eucommiae cortex, which is used for treating arthritis, osteoporotic fractures, and hypertension. However, the therapeutic effects of GPA against osteoporosis remain underexplored. PURPOSE This study aims to reveal the potential osteogenic effects of FXR and to explore the effect of GPA on bone formation, osteoporosis treatment, and FXR signaling. STUDY DESIGN & METHODS The role of FXR in promoting bone formation was evaluated in Fxr knockout (Fxr-/-) mice and cell models. GPA activation of FXR was evaluated by molecular docking and luciferase reporter gene assays. Thirty female C57BL/6J mice were randomly assigned into a sham operation group (Sham) and four ovariectomized (OVX) groups (n=6 each) and were treated with vehicle or different doses of GPA (25, 50, and 100 mg/kg/day). The therapeutic effect of GPA on osteoporosis was systematically analyzed by performing bone histomorphometry and measuring serum biochemical parameters, and the molecular mechanism was also evaluated. Furthermore, the action of GPA in Fxr-/- mice was evaluated to investigate its dependency on FXR in promoting bone formation and treating osteoporosis. RESULTS We found that FXR was highly expressed in bone tissues and enriched in osteoblasts. Notably, deletion of FXR significantly reduced the bone formation rate and bone mass of the Fxr-/- mice compared with wild-type mice. Furthermore, using a high throughput drug screening strategy based on fluorescent reporter genes, we found that GPA functions as a natural agonist of FXR. We confirmed the activities of GPA on FXR activation and osteogenesis in both osteoblast differentiation models and OVX-induced osteoporosis models. We revealed that GPA strongly promotes bone formation by activating FXR/RUNX2 signaling. Moreover, the osteoporotic therapeutic effect of GPA was abolished in Fxr-/- mice. CONCLUSION This study demonstrated that FXR is a promising target for treating osteoporosis and that GPA promotes bone formation in OVX-induced osteoporosis by activating FXR signaling. These findings provide novel insight into the mechanism by which GPA promotes bone formation and more evidence for its application in the treatment of osteoporosis.
Collapse
Affiliation(s)
- Meijing Liu
- Key Laboratory of Big Data-Based Precision Medicine, School of Engineering Medicine, Beihang University, Beijing, 100191, China; Clinical Research Platform for Interdiscipline of Stomatology, The First Affiliated Hospital of Jinan University & Department of Stomatology, Jinan University, Guangzhou, 510632, China; School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Fujun Jin
- Key Laboratory of Big Data-Based Precision Medicine, School of Engineering Medicine, Beihang University, Beijing, 100191, China
| | - Shuai Zhang
- Department of Oral Implantology, School of Stomatology, Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, 200072, China
| | - Shuang Li
- Key Laboratory of Big Data-Based Precision Medicine, School of Engineering Medicine, Beihang University, Beijing, 100191, China
| | - Danqi Zhu
- Key Laboratory of Big Data-Based Precision Medicine, School of Engineering Medicine, Beihang University, Beijing, 100191, China
| | - Yi Cui
- Key Laboratory of Big Data-Based Precision Medicine, School of Engineering Medicine, Beihang University, Beijing, 100191, China
| | - Mingxiang Cai
- Clinical Research Platform for Interdiscipline of Stomatology, The First Affiliated Hospital of Jinan University & Department of Stomatology, Jinan University, Guangzhou, 510632, China
| | - Xiangning Liu
- Clinical Research Platform for Interdiscipline of Stomatology, The First Affiliated Hospital of Jinan University & Department of Stomatology, Jinan University, Guangzhou, 510632, China
| | - Yongbiao Zhang
- Key Laboratory of Big Data-Based Precision Medicine, School of Engineering Medicine, Beihang University, Beijing, 100191, China
| | - Yao Sun
- Department of Oral Implantology, School of Stomatology, Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, 200072, China.
| | - Changhui Liu
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China..
| | - Xiaogang Wang
- Key Laboratory of Big Data-Based Precision Medicine, School of Engineering Medicine, Beihang University, Beijing, 100191, China; Clinical Research Platform for Interdiscipline of Stomatology, The First Affiliated Hospital of Jinan University & Department of Stomatology, Jinan University, Guangzhou, 510632, China.
| |
Collapse
|
21
|
Composite Coatings for Osteoblast Growth Attachment Fabricated by Matrix-Assisted Pulsed Laser Evaporation. Polymers (Basel) 2022; 14:polym14142934. [PMID: 35890714 PMCID: PMC9322700 DOI: 10.3390/polym14142934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 07/14/2022] [Accepted: 07/18/2022] [Indexed: 11/16/2022] Open
Abstract
The bioactive and biocompatible properties of hydroxyapatite (HAp) promote the osseointegration process. HAp is widely used in biomedical applications, especially in orthopedics, as well as a coating material for metallic implants. We obtained composite coatings based on HAp, chitosan (CS), and FGF2 by a matrix-assisted pulsed laser evaporation (MAPLE) technique. The coatings were physico-chemically investigated by means of X-ray Diffraction (XRD), Transmission Electron Microscopy (TEM), Infrared Microscopy (IRM), and Scanning Electron Microscopy (SEM). Further, biological investigations were performed. The MAPLE-composite coatings were tested in vitro on the MC3T3-E1 cell line in order to endorse cell attachment and growth without toxic effects and to promote pre-osteoblast differentiation towards the osteogenic lineage. These coatings can be considered suitable for bone tissue engineering applications that lack toxicity and promotes cell adhesion and proliferation while also sustaining the differentiation of pre-osteoblasts towards mature bone cells.
Collapse
|
22
|
Fois MG, Tahmasebi Birgani ZN, Guttenplan APM, Blitterswijk CAV, Giselbrecht S, Habibović P, Truckenmüller RK. Assessment of Cell-Material Interactions in Three Dimensions through Dispersed Coaggregation of Microsized Biomaterials into Tissue Spheroids. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2202112. [PMID: 35754160 DOI: 10.1002/smll.202202112] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 05/23/2022] [Indexed: 06/15/2023]
Abstract
In biomaterials R&D, conventional monolayer cell culture on flat/planar material samples, such as films, is still commonly employed at early stages of the assessment of interactions of cells with candidate materials considered for a biomedical application. In this feasibility study, an approach for the assessment of 3D cell-material interactions through dispersed coaggregation of microparticles from biomaterials into tissue spheroids is presented. Biomaterial microparticles can be created comparatively quickly and easily, allow the miniaturization of the assessment platform, and enable an unhindered remodeling of the dynamic cell-biomaterial system at any time. The aggregation of the microsized biomaterials and the cells is supported by low-attachment round-bottom microwells from thin polymer films arranged in densely packed arrays. The study is conducted by the example of MG63 osteoblast-like and human mesenchymal stem/stromal cells, and a small library of model microbiomaterials related to bone repair and regeneration. For the proof of concept, example interactions including cell adhesion to the material, the hybrid spheroids' morphology, size, and shape, material-associated cell death, cell metabolic activity, cell proliferation, and (osteogenic) differentiation are investigated. The cells in the spheroids are shown to respond to differences in the microbiomaterials' properties, their amounts, and the duration of interaction with them.
Collapse
Affiliation(s)
- Maria G Fois
- MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, P.O. Box 616, Maastricht, MD, 6200, The Netherlands
| | - Zeinab N Tahmasebi Birgani
- MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, P.O. Box 616, Maastricht, MD, 6200, The Netherlands
| | - Alexander P M Guttenplan
- MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, P.O. Box 616, Maastricht, MD, 6200, The Netherlands
| | - Clemens A van Blitterswijk
- MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, P.O. Box 616, Maastricht, MD, 6200, The Netherlands
| | - Stefan Giselbrecht
- MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, P.O. Box 616, Maastricht, MD, 6200, The Netherlands
| | - Pamela Habibović
- MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, P.O. Box 616, Maastricht, MD, 6200, The Netherlands
| | - Roman K Truckenmüller
- MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, P.O. Box 616, Maastricht, MD, 6200, The Netherlands
| |
Collapse
|
23
|
Khokhani P, Belluomo R, Croes M, Gawlitta D, Kruyt MC, Weinans H. An in-vitro model to test the influence of immune cell secretome on MSC osteogenic differentiation. Tissue Eng Part C Methods 2022; 28:420-430. [PMID: 35770885 DOI: 10.1089/ten.tec.2022.0086] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Immune cells and their soluble factors have an important role in the bone healing process. Modulation of the immune response, therefore, offers a potential strategy to enhance bone formation. To investigate the influence of the immune system on osteogenesis, we developed and applied an in-vitro model that incorporates both innate and adaptive immune cells. Human peripheral blood mononuclear cells (PBMCs) were isolated and cultured for 24 hours and subsequently stimulated with immune-modulatory agents; C-class CpG oligodeoxynucleotide (CpG ODN C), Polyinosinic acid-polycytidylic acid Poly(I:C), and lipopolysaccharide (LPS); all pathogen recognition receptor agonists, and that target Toll-like receptors TLR9, -3, and -4, respectively. The conditioned medium obtained from PBMCs after 24 hours was used to investigate its effects on the metabolic activity and osteogenic differentiation capacity of human bone marrow-derived mesenchymal stromal cells (MSCs). Conditioned media from unstimulated PBMCs did not affect the metabolic activity and osteogenic differentiation capacity of MSCs. The conditioned medium from CpG ODN C and LPS stimulated PBMCs increased alkaline phosphatase activity of MSCs by approximately 3-fold as compared to the unstimulated control, whereas Poly(I:C) conditioned medium did not enhance ALP activity of MSCs. Moreover, direct stimulation of MSCs with the immune-modulatory stimuli did not result in increased alkaline phosphatase activity. These results demonstrate that soluble factors present in conditioned medium from PBMCs stimulated with immune-modulatory factors enhance osteogenesis of MSCs. This in-vitro model can serve as a tool in screening immune-modulatory stimulants from a broad variety of immune cells for (indirect) effects on osteogenesis and also to identify soluble factors from multiple immune cell types that may modulate bone healing.
Collapse
Affiliation(s)
- Paree Khokhani
- University Medical Centre Utrecht, 8124, Orthopedics , UMC Utrecht, dept. Orthopedics, G5.203, Heidelberglaan 100, Utrecht, Utrecht, Drenthe, Netherlands, 3584CX.,University Medical Centre, Utrecht (UMCU), UMC Utrecht, dept. Orthopedics, G5.203, Heidelberglaan 100, Netherlands;
| | - Ruggero Belluomo
- University Medical Centre Utrecht, 8124, Orthopedics , Utrecht, Utrecht, Netherlands;
| | - Michiel Croes
- University Medical Centre Utrecht, 8124, Orthopedics , Utrecht, Utrecht, Netherlands;
| | - Debby Gawlitta
- University Medical Center Utrecht, Oral and Maxillofacial Surgery, Prosthodontics & Special Dental Care, Heidelberglaan 100, G05.129, PO Box 85500, Utrecht, Netherlands, 3508 GA;
| | - Moyo C Kruyt
- University medical center Utrecht, Orthopedics, HP G 05.228, PO Box 85500, Utrecht, Netherlands, 3508 GA;
| | - Harrie Weinans
- University Medical Centre Utrecht, 8124, Orthopedics, Utrecht, Utrecht, Netherlands;
| |
Collapse
|
24
|
Bogdanova M, Zabirnyk A, Malashicheva A, Semenova D, Kvitting JPE, Kaljusto ML, Perez MDM, Kostareva A, Stensløkken KO, Sullivan GJ, Rutkovskiy A, Vaage J. Models and Techniques to Study Aortic Valve Calcification in Vitro, ex Vivo and in Vivo. An Overview. Front Pharmacol 2022; 13:835825. [PMID: 35721220 PMCID: PMC9203042 DOI: 10.3389/fphar.2022.835825] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 04/29/2022] [Indexed: 11/23/2022] Open
Abstract
Aortic valve stenosis secondary to aortic valve calcification is the most common valve disease in the Western world. Calcification is a result of pathological proliferation and osteogenic differentiation of resident valve interstitial cells. To develop non-surgical treatments, the molecular and cellular mechanisms of pathological calcification must be revealed. In the current overview, we present methods for evaluation of calcification in different ex vivo, in vitro and in vivo situations including imaging in patients. The latter include echocardiography, scanning with computed tomography and magnetic resonance imaging. Particular emphasis is on translational studies of calcific aortic valve stenosis with a special focus on cell culture using human primary cell cultures. Such models are widely used and suitable for screening of drugs against calcification. Animal models are presented, but there is no animal model that faithfully mimics human calcific aortic valve disease. A model of experimentally induced calcification in whole porcine aortic valve leaflets ex vivo is also included. Finally, miscellaneous methods and aspects of aortic valve calcification, such as, for instance, biomarkers are presented.
Collapse
Affiliation(s)
- Maria Bogdanova
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Arsenii Zabirnyk
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway.,Department of Research and Development, Division of Emergencies and Critical Care, Oslo University Hospital, Oslo, Norway
| | - Anna Malashicheva
- Institute of Cytology, Russian Academy of Sciences, Saint Petersburg, Russia
| | - Daria Semenova
- Institute of Cytology, Russian Academy of Sciences, Saint Petersburg, Russia
| | | | - Mari-Liis Kaljusto
- Department of Cardiothoracic Surgery, Oslo University Hospital, Oslo, Norway
| | | | - Anna Kostareva
- Almazov National Medical Research Centre, Saint Petersburg, Russia.,Department of Woman and Children Health, Karolinska Institute, Stockholm, Sweden
| | - Kåre-Olav Stensløkken
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Gareth J Sullivan
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway.,Norwegian Center for Stem Cell Research, Oslo University Hospital and University of Oslo, Oslo, Norway.,Institute of Immunology, Oslo University Hospital, Oslo, Norway.,Hybrid Technology Hub - Centre of Excellence, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway.,Department of Pediatric Research, Oslo University Hospital, Oslo, Norway
| | - Arkady Rutkovskiy
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway.,Department of Pulmonary Diseases, Oslo University Hospital, Oslo, Norway
| | - Jarle Vaage
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway.,Department of Research and Development, Division of Emergencies and Critical Care, Oslo University Hospital, Oslo, Norway.,Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
25
|
Discovery of surface biomarkers for cell mechanophenotype via an intracellular protein-based enrichment strategy. Cell Mol Life Sci 2022; 79:320. [PMID: 35622146 DOI: 10.1007/s00018-022-04351-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 04/28/2022] [Accepted: 05/05/2022] [Indexed: 11/03/2022]
Abstract
Cellular mechanophenotype is often a defining characteristic of conditions like cancer malignancy/metastasis, cardiovascular disease, lung and liver fibrosis, and stem cell differentiation. However, acquiring living cells based on mechanophenotype is challenging for conventional cell sorters due to a lack of biomarkers. In this study, we demonstrate a workflow for surface protein discovery associated with cellular mechanophenotype. We sorted heterogeneous adipose-derived stem/stromal cells (ASCs) into groups with low vs. high lamin A/C, an intracellular protein linked to whole-cell mechanophenotype. Proteomic data of enriched groups identified surface protein candidates as potential biochemical proxies for ASC mechanophenotype. Select surface biomarkers were used for live-cell enrichment, with subsequent single-cell mechanical testing and lineage-specific differentiation. Ultimately, we identified CD44 to have a strong inverse correlation with whole-cell elastic modulus, with CD44lo cells exhibiting moduli three times greater than that of CD44hi cells. Functionally, these stiff and soft ASCs showed enhanced osteogenic and adipogenic differentiation potential, respectively. The described workflow can be replicated for any phenotype with a known correlated intracellular protein, allowing for the acquisition of live cells for further characterization, diagnostics, or therapeutics.
Collapse
|
26
|
Hegedűs C, Czibulya Z, Tóth F, Dezső B, Hegedűs V, Boda R, Horváth D, Csík A, Fábián I, Tóth-Győri E, Sajtos Z, Lázár I. The Effect of Heat Treatment of β-Tricalcium Phosphate-Containing Silica-Based Bioactive Aerogels on the Cellular Metabolism and Proliferation of MG63 Cells. Biomedicines 2022; 10:662. [PMID: 35327463 PMCID: PMC8945762 DOI: 10.3390/biomedicines10030662] [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: 02/08/2022] [Revised: 02/22/2022] [Accepted: 03/10/2022] [Indexed: 02/06/2023] Open
Abstract
β-Tricalcium phosphate was combined with silica aerogel in composites prepared using the sol-gel technique and supercritical drying. The materials were used in this study to check their biological activity and bone regeneration potential with MG63 cell experiments. The composites were sintered in 100 °C steps in the range of 500-1000 °C. Their mechanical properties, porosities, and solubility were determined as a function of sintering temperature. Dissolution studies revealed that the released Ca-/P molar ratios appeared to be in the optimal range to support bone tissue induction. Cell viability, ALP activity, and type I collagen gene expression results all suggested that the sintering of the compound at approximately 700-800 °C as a scaffold could be more powerful in vivo to facilitate bone formation within a bone defect, compared to that documented previously by our research team. We did not observe any detrimental effect on cell viability. Both the alkaline phosphatase enzyme activity and the type I collagen gene expression were significantly higher compared with the control and the other aerogels heat-treated at different temperatures. The mesoporous silica-based aerogel composites containing β-tricalcium phosphate particles treated at temperatures lower than 1000 °C produced a positive effect on the osteoblastic activity of MG63 cells. An in vivo 6 month-long follow-up study of the mechanically strongest 1000 °C sample in rat calvaria experiments provided proof of a complete remodeling of the bone.
Collapse
Affiliation(s)
- Csaba Hegedűs
- Department of Biomaterials and Prosthetic Dentistry, Faculty of Dentistry, University of Debrecen, 4032 Debrecen, Hungary; (Z.C.); (F.T.)
| | - Zsuzsanna Czibulya
- Department of Biomaterials and Prosthetic Dentistry, Faculty of Dentistry, University of Debrecen, 4032 Debrecen, Hungary; (Z.C.); (F.T.)
| | - Ferenc Tóth
- Department of Biomaterials and Prosthetic Dentistry, Faculty of Dentistry, University of Debrecen, 4032 Debrecen, Hungary; (Z.C.); (F.T.)
| | - Balázs Dezső
- Department of Oral Pathology and Microbiology, Faculty of Dentistry, University of Debrecen, 4032 Debrecen, Hungary;
| | - Viktória Hegedűs
- Department of Pediatric Dentistry and Orthodontics, Faculty of Dentistry, University of Debrecen, 4032 Debrecen, Hungary;
| | - Róbert Boda
- Department of Oral and Maxillofacial Surgery, Faculty of Dentistry, University of Debrecen, 4032 Debrecen, Hungary; (R.B.); (D.H.)
| | - Dóra Horváth
- Department of Oral and Maxillofacial Surgery, Faculty of Dentistry, University of Debrecen, 4032 Debrecen, Hungary; (R.B.); (D.H.)
| | - Attila Csík
- Laboratory of Materials Science, Institute for Nuclear Research, Eötvös Loránd Research Network, 4026 Debrecen, Hungary;
| | - István Fábián
- Department of Inorganic and Analytical Chemistry, Faculty of Science and Technology, University of Debrecen, 4032 Debrecen, Hungary; (I.F.); (E.T.-G.); (Z.S.)
| | - Enikő Tóth-Győri
- Department of Inorganic and Analytical Chemistry, Faculty of Science and Technology, University of Debrecen, 4032 Debrecen, Hungary; (I.F.); (E.T.-G.); (Z.S.)
| | - Zsófi Sajtos
- Department of Inorganic and Analytical Chemistry, Faculty of Science and Technology, University of Debrecen, 4032 Debrecen, Hungary; (I.F.); (E.T.-G.); (Z.S.)
| | - István Lázár
- Department of Inorganic and Analytical Chemistry, Faculty of Science and Technology, University of Debrecen, 4032 Debrecen, Hungary; (I.F.); (E.T.-G.); (Z.S.)
| |
Collapse
|
27
|
Mineralizing Gelatin Microparticles as Cell Carrier and Drug Delivery System for siRNA for Bone Tissue Engineering. Pharmaceutics 2022; 14:pharmaceutics14030548. [PMID: 35335924 PMCID: PMC8949427 DOI: 10.3390/pharmaceutics14030548] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 02/24/2022] [Accepted: 02/25/2022] [Indexed: 11/19/2022] Open
Abstract
The local release of complexed siRNA from biomaterials opens precisely targeted therapeutic options. In this study, complexed siRNA was loaded to gelatin microparticles cross-linked (cGM) with an anhydride-containing oligomer (oPNMA). We aggregated these siRNA-loaded cGM with human mesenchymal stem cells (hMSC) to microtissues and stimulated them with osteogenic supplements. An efficient knockdown of chordin, a BMP-2 antagonist, caused a remarkably increased alkaline phosphatase (ALP) activity in the microtissues. cGM, as a component of microtissues, mineralized in a differentiation medium within 8–9 days, both in the presence and in the absence of cells. In order to investigate the effects of our pre-differentiated and chordin-silenced microtissues on bone homeostasis, we simulated in vivo conditions in an unstimulated co-culture system of hMSC and human peripheral blood mononuclear cells (hPBMC). We found enhanced ALP activity and osteoprotegerin (OPG) secretion in the model system compared to control microtissues. Our results suggest osteoanabolic effects of pre-differentiated and chordin-silenced microtissues.
Collapse
|
28
|
Touya N, Devun M, Handschin C, Casenave S, Omar NA, Gaubert A, Dusserre N, De Oliveira H, Kérourédan O, Devillard R. In vitroand in vivocharacterization of a novel tricalcium silicate-based ink for bone regeneration using laser-assisted bioprinting. Biofabrication 2022; 14. [PMID: 35203068 DOI: 10.1088/1758-5090/ac584b] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 02/24/2022] [Indexed: 11/11/2022]
Abstract
Grafts aside, current strategies employed to overcome bone loss still fail to reproduce native tissue physiology. Among the emerging bioprinting strategies, Laser-Assisted Bioprinting (LAB) offers very high resolution, allowing designing micrometric patterns in a contactless manner, providing a reproducible tool to test ink formulation. To this date, no LAB associated ink succeeded to provide a reproducible ad integrum bone regeneration on a murine calvaria critical size defect model. Using the CE approved BioRoot RCS® as a mineral addition to a collagen-enriched ink compatible with LAB, the present study describes the process of the development of a solidifying tricalcium silicate-based ink as a new bone repair promoting substrates in a LAB model. This ink formulation was mechanically characterized by rheology to adjust it for LAB. Printed aside Stromal Cells from Apical Papilla (SCAPs), this ink demonstrated a great cytocompatibility, with significant in vitro positive impact upon cell motility, and an early osteogenic differentiation response in the absence of another stimulus. Results indicated that the in vivo application of this new ink formulation to regenerate critical size bone defect tends to promote the formation of bone volume fraction without affecting the vascularization of the neo-formed tissue. The use of LAB techniques with this ink failed to demonstrate a complete bone repair, whether SCAPs were printed or not of at its direct proximity. The relevance of the properties of this specific ink formulation would therefore rely on the quantity applied in situ as a defect filler rather than its cell modulation properties observed in vitro. For the first time, a tricalcium silicate-based printed ink, based on rheological analysis, was characterized in vitro and in vivo, giving valuable information to reach complete bone regeneration through formulation updates. This LAB-based process could be generalized to normalize the characterization of candidate ink for bone regeneration.
Collapse
Affiliation(s)
- Nicolas Touya
- University of Bordeaux, 146 rue leo saignat, Bordeaux, 33067, FRANCE
| | - Mathilde Devun
- University of Bordeaux, 146 rue leo saignat, Talence, 33405, FRANCE
| | - Charles Handschin
- Inserm U1026, Tissue Bioengineering: Bordeaux, FR, 146 rue leo saignat, Bordeaux, 33067, FRANCE
| | - Sophia Casenave
- University of Bordeaux, 146 rue leo saignat, Talence, 33405, FRANCE
| | - Naïma Ahmed Omar
- University of Bordeaux, 146 rue leo saignat, Talence, 33405, FRANCE
| | - Alexandra Gaubert
- University of Bordeaux, 146 rue leo saignat, Bordeaux, 33067, FRANCE
| | - Nathalie Dusserre
- ART Bioprint, INSERM U1026, 146 rue leo saignat, BORDEAUX, 33067, FRANCE
| | - Hugo De Oliveira
- , Université de Bordeaux, Bioingénierie tissulaire, rue Léo Saignat, 33076 Bordeaux, Bordeaux, 33067, FRANCE
| | - Olivia Kérourédan
- Bioingénierie Tissulaire, INSERM U1026, 146 rue Léo Saignat, BORDEAUX, 33067, FRANCE
| | - Raphael Devillard
- Bioingenierie tissulaire, INSERM U1026, 146 rue leo Saignat, Bordeaux, 33067, FRANCE
| |
Collapse
|
29
|
Ambattu LA, Gelmi A, Yeo LY. Short-Duration High Frequency MegaHertz-Order Nanomechanostimulation Drives Early and Persistent Osteogenic Differentiation in Mesenchymal Stem Cells. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2106823. [PMID: 35023629 DOI: 10.1002/smll.202106823] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Revised: 12/12/2021] [Indexed: 06/14/2023]
Abstract
Stem cell fate can be directed through the application of various external physical stimuli, enabling a controlled approach to targeted differentiation. Studies involving the use of dynamic mechanical cues driven by vibrational excitation to date have, however, been limited to low frequency (Hz to kHz) forcing over extended durations (typically continuous treatment for >7 days). Contrary to previous assertions that there is little benefit in applying frequencies beyond 1 kHz, we show here that high frequency MHz-order mechanostimulation in the form of nanoscale amplitude surface reflected bulk waves are capable of triggering differentiation of human mesenchymal stem cells from various donor sources toward an osteoblast lineage, with early, short time stimuli inducing long-term osteogenic commitment. More specifically, rapid treatments (10 min daily over 5 days) of the high frequency (10 MHz) mechanostimulation are shown to trigger significant upregulation in early osteogenic markers (RUNX2, COL1A1) and sustained increase in late markers (osteocalcin, osteopontin) through a mechanistic pathway involving piezo channel activation and Rho-associated protein kinase signaling. Given the miniaturizability and low cost of the devices, the possibility for upscaling the platform toward practical bioreactors, to address a pressing need for more efficient stem cell differentiation technologies in the pursuit of translatable regenerative medicine strategies, is ensivaged.
Collapse
Affiliation(s)
- Lizebona August Ambattu
- Micro/Nanophysics Research Laboratory, School of Engineering, RMIT University, Melbourne, Victoria, 3000, Australia
| | - Amy Gelmi
- School of Science, RMIT University, Melbourne, Victoria, 3000, Australia
| | - Leslie Y Yeo
- Micro/Nanophysics Research Laboratory, School of Engineering, RMIT University, Melbourne, Victoria, 3000, Australia
| |
Collapse
|
30
|
Skiba G, Raj S, Sobol M, Kowalczyk P, Barszcz M, Taciak M, Tuśnio A, Čobanová K, Grešáková Ľ, Grela ER. Influence of the Zinc and Fibre Addition in the Diet on Biomechanical Bone Properties in Weaned Piglets. Animals (Basel) 2022; 12:181. [PMID: 35049803 PMCID: PMC8773129 DOI: 10.3390/ani12020181] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 01/05/2022] [Accepted: 01/10/2022] [Indexed: 12/31/2022] Open
Abstract
The effects of the zinc and fibre source in piglets' diet on the bone mineral content, density, and strength parameters of the femur were investigated using 24 piglets fed a diet supplemented with either lignocellulose (LC) or potato fibre (PF). Half of each group of piglets consumed a diet with ZnSO4 monohydrate or with zinc glycinate (ZnGly). The diets contained similar amounts of lysine, energy, and fibre. Bone mineral content and density were over 9% higher in pigs receiving diets with ZnGly than in animals fed diets with ZnSO4. Moreover, ZnGly strongly improved maximum and elastic strength (by 25.7 and 20.0%, respectively, p < 0.0001) and bone stiffness (by 29.4%, p < 0.0001). Only the mass of the femur was affected by the type of fibre in the diet, as the femurs of piglets fed diets with LC were over 7% (p < 0.0001) heavier than in piglets fed diets with PF. The intake of digestible zinc and the zinc content in the blood serum were positively correlated with the measured bone parameters and, depending on the parameter, "r" ranged from 0.749 to 0.866 and from 0.400 to 0.479, respectively. It can be concluded that bone parameters are affected more strongly by the organic than inorganic source of zinc.
Collapse
Affiliation(s)
- Grzegorz Skiba
- The Kielanowski Institute of Animal Physiology and Nutrition, Department of Animal Nutrition, Polish Academy of Sciences, Instytucka 3, 05-110 Jabłonna, Poland; (S.R.); (M.S.); (P.K.); (M.B.); (M.T.); (A.T.)
| | - Stanisława Raj
- The Kielanowski Institute of Animal Physiology and Nutrition, Department of Animal Nutrition, Polish Academy of Sciences, Instytucka 3, 05-110 Jabłonna, Poland; (S.R.); (M.S.); (P.K.); (M.B.); (M.T.); (A.T.)
| | - Monika Sobol
- The Kielanowski Institute of Animal Physiology and Nutrition, Department of Animal Nutrition, Polish Academy of Sciences, Instytucka 3, 05-110 Jabłonna, Poland; (S.R.); (M.S.); (P.K.); (M.B.); (M.T.); (A.T.)
| | - Paweł Kowalczyk
- The Kielanowski Institute of Animal Physiology and Nutrition, Department of Animal Nutrition, Polish Academy of Sciences, Instytucka 3, 05-110 Jabłonna, Poland; (S.R.); (M.S.); (P.K.); (M.B.); (M.T.); (A.T.)
| | - Marcin Barszcz
- The Kielanowski Institute of Animal Physiology and Nutrition, Department of Animal Nutrition, Polish Academy of Sciences, Instytucka 3, 05-110 Jabłonna, Poland; (S.R.); (M.S.); (P.K.); (M.B.); (M.T.); (A.T.)
| | - Marcin Taciak
- The Kielanowski Institute of Animal Physiology and Nutrition, Department of Animal Nutrition, Polish Academy of Sciences, Instytucka 3, 05-110 Jabłonna, Poland; (S.R.); (M.S.); (P.K.); (M.B.); (M.T.); (A.T.)
| | - Anna Tuśnio
- The Kielanowski Institute of Animal Physiology and Nutrition, Department of Animal Nutrition, Polish Academy of Sciences, Instytucka 3, 05-110 Jabłonna, Poland; (S.R.); (M.S.); (P.K.); (M.B.); (M.T.); (A.T.)
| | - Klaudia Čobanová
- Institute of Animal Physiology, Centre of Biosciences of the Slovak Academy of Sciences, Soltesovej 4, 04-001 Košice, Slovakia; (K.Č.); (Ľ.G.)
| | - Ľubomira Grešáková
- Institute of Animal Physiology, Centre of Biosciences of the Slovak Academy of Sciences, Soltesovej 4, 04-001 Košice, Slovakia; (K.Č.); (Ľ.G.)
| | - Eugeniusz Ryszard Grela
- Institute of Animal Nutrition and Bromatology, University of Life Sciences in Lublin, Akademicka 13, 20-950 Lublin, Poland;
| |
Collapse
|
31
|
Alghfeli L, Parambath D, Manzoor S, Roach HI, Oreffo RO, El-Serafi AT. Synthesis of scaffold-free, three dimensional, osteogenic constructs following culture of skeletal osteoprogenitor cells on glass surfaces. Bone Rep 2021; 15:101143. [PMID: 34746337 PMCID: PMC8554168 DOI: 10.1016/j.bonr.2021.101143] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 10/06/2021] [Accepted: 10/08/2021] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Efficient differentiation of stem cells into three-dimensional (3D) osteogenic construct is still an unmet challenge. These constructs can be crucial for patients with bone defects due to congenital or traumatic reasons. The modulation of cell fate and function as a consequence of interaction with the physical and chemical properties of materials is well known. METHODS The current study has examined the osteogenic differentiation potential of human skeletal populations following culture on glass surfaces, as a monolayer, or in glass tubes as a pellet culture. The 3D prosperities were assessed morphometrically and the differentiation was evaluated through molecular characterization as well as matrix formation. RESULTS Early temporal expression of alkaline phosphatase expression of skeletal populations was observed following culture on glass surfaces. Skeletal populations seeded on glass tubes, adhered as a monolayer to the tube base and subsequently formed 3D pellets at the air -media interface. The pellets cultured on glass displayed 4.9 ± 1.3 times the weight and 2.9 ± 0.1 the diameter of their counterpart cultured in plastic tubes and displayed enhanced production of osteogenic matrix proteins, such a collagen I and osteonectin. The size and weight of the pellets correlated with surface area in contrast to cell numbers seeded. Global DNA methylation level was decreased in pellets cultured on glass. In contrast, gene expression analysis confirmed upregulation extracellular matrix proteins and osteogenesis-related growth factors. CONCLUSION This simple approach to the culture of skeletal cells on glass tubes provides a scaffold-free, 3D construct platform for generating pellets enabling analysis and evaluation of tissue development and integration of multiple constructs with implications for tissue repair and regenerative application on scale-up.
Collapse
Key Words
- 3D culture
- 3D, three-dimensional
- A/S, Alcian blue/Sirius red/Weigert's haematoxylin
- ALP, Alkaline Phosphatase
- BMP, bone morphogenic protein
- BMSC, human bone marrow stromal
- CSF, colony stimulating factor
- Ct, Cycle threshold
- Differentiation
- EGF, epidermal growth factor
- FC, Fetal bone cells
- FCS, Fetal Calf Serum
- FGF, fibroblast growth factor
- FN1, fibronectin
- GLI, GLI family zinc finger 1
- HIPPIE, Human Integrated Protein Interaction Reference
- ITGA3, integrin A3
- MMP, matrix metalloprotease
- Osteogenesis
- Osteoprogenitor cells
- P/S, penicillin and streptomycin
- Pellets
- R, receptor
- TGF, β transforming growth factor beta
- TGFBR2 transforming growth factor beta receptor 2 VDR, vitamin D receptor
- gDNA, genomic DNA
- iMSC, immortalized human bone marrow derived, mesenchymal stem cells
- vWF, von Willebrand factor
Collapse
Affiliation(s)
- Latifa Alghfeli
- Research Institute for Medical and Health Sciences, University of Sharjah, United Arab Emirates
| | - Divyasree Parambath
- Research Institute for Medical and Health Sciences, University of Sharjah, United Arab Emirates
| | - Shaista Manzoor
- Research Institute for Medical and Health Sciences, University of Sharjah, United Arab Emirates
| | - Helmtrud I. Roach
- Bone and Joint Research Group, Institute of Developmental Sciences, University of Southampton, School of Medicine, UK
| | - Richard O.C. Oreffo
- Bone and Joint Research Group, Institute of Developmental Sciences, University of Southampton, School of Medicine, UK
| | - Ahmed T. El-Serafi
- Research Institute for Medical and Health Sciences, University of Sharjah, United Arab Emirates
- Medical Biochemistry Department, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
- Department of Biomedical and Clinical Sciences (BKV), Linköping University, Sweden
| |
Collapse
|
32
|
Sun X, Yu X, Li W, Chen M, Liu D. Fabrication and characterization of biodegradable zinc matrix composites reinforced by uniformly dispersed beta-tricalcium phosphate via graphene oxide-assisted hetero-agglomeration. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 130:112431. [PMID: 34702516 DOI: 10.1016/j.msec.2021.112431] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Revised: 09/03/2021] [Accepted: 09/08/2021] [Indexed: 10/20/2022]
Abstract
The development of biodegradable Zn matrix composites has been considered a promising approach to achieving enhanced mechanical properties, controllable degradation rate, good biocompatibility, and good osseointegration as orthopedic implants. However, scant literature regarding Zn matrix composites has been reported because of the great difficulty in dispersing the nano-sized bioactive reinforcements uniformly within the Zn matrix. In the present study, a novel and effective method were employed to obtain Zn matrix composites reinforced by uniformly dispersed beta-tricalcium phosphate (β-TCP) via graphene oxide (GO)-assisted hetero-agglomeration and subsequent spark plasma sintering process. A very low-content (0.04 vol%) few-layered GO was used as a coupling reagent to connect the Zn matrix and nano-sized TCP particles. In an appropriate polarity solvent, the negatively charged GO sheets could combine with both the positively charged Zn powder and TCP particles by electrostatic attraction and charge neutralization. Due to the nature of hetero-agglomeration, the flexible GO sheet could adhere to the large Zn powder and attracted a certain amount of TCP particles to form a Zn/GO/TCP sandwich structure by charge neutralization thereby forming a uniform dispersion of TCP particles within Zn matrix. After the spark plasma sintering (SPS) process, the TCP particles incorporated with very thin ZnO layers (thickness of a few dozen nanometers) formed a homogeneous and unique 3D network-like distribution in as-sintered TCP/Zn composites. A unique "snap pea"-like structure was confirmed at the grain boundary of α-Zn grains, which consisted of the TCP particles as "pea" and thin ZnO layer as "pod". Due to the uniform dispersion of bioactive TCP particles and unique structure of the TCP incorporating grain boundary, as-sintered 3TCP/Zn matrix composites possessed yield strength (YS) of 140.8 ± 7.7 MPa, failure strain of 36.0 ± 2.8%, the moderate degradation rate of 19.1 ± 3.3 μm·y-1 and good cytocompatibility to MC3T3-E1 cells. Moreover, osteogenic differentiation activity evaluation revealed that the addition of TCP could significantly improve the expressions of the osteogenic differentiation-related gene (ALP) in MC3T3-E1 cells, thereby resulting in improved osteogenic capability. Therefore, biodegradable 3TCP/Zn matrix composites fabricated by GO-assisted hetero-agglomeration and subsequent SPS process could be a promising material as orthopedic implants.
Collapse
Affiliation(s)
- Xiaohao Sun
- School of Materials Science and Engineering, Tianjin University of Technology, Tianjin 300384, China.
| | - Xiao Yu
- School of Materials Science and Engineering, Tianjin University of Technology, Tianjin 300384, China
| | - Wei Li
- School of Chemistry and Chemical Engineering, Tianjin University of Technology, Tianjin 300384, China
| | - Minfang Chen
- National Demonstration Center for Experimental Function Materials Education, Tianjin University of Technology, Tianjin 300384, China
| | - Debao Liu
- Tianjin Key Laboratory for Photoelectric Materials and Devices, Tianjin 300384, China.
| |
Collapse
|
33
|
Micro-Computed Tomography Analysis on Administration of Mesenchymal Stem Cells - Bovine Teeth Scaffold Composites for Alveolar Bone Tissue Engineering. JOURNAL OF BIOMIMETICS BIOMATERIALS AND BIOMEDICAL ENGINEERING 2021. [DOI: 10.4028/www.scientific.net/jbbbe.52.86] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The tissue engineering approach for periodontal tissue regeneration using a combination of stem cells and scaffold has been vastly developed. Mesenchymal Stem Cells (MSCs) seeded with Bovine Teeth Scaffold (BTSc) can repair alveolar bone damage in periodontitis cases. The alveolar bone regeneration process was analyzed by micro-computed tomography (µ-CT) to observe the structure of bone growth and to visualize the scaffold in 3-Dimensional (3D). The purpose of this study is to analyze alveolar bone regeneration by µ-CT following the combination of MSCs and bovine teeth scaffold (MSCs-BTSc) implantation in the Wistar rat periodontitis model. Methods. MSCs were cultured from adipose-derived mesenchymal stem cells of rats. BTSc was taken from bovine teeth and freeze-dried with a particle size of 150-355 µm. MSCs were seeded on BTSc for 24 hours and transplanted in a rat model of periodontitis. Thirty-five Wistar rats were made as periodontitis models with LPS induction from P. gingivalis injected to the buccal section of interproximal gingiva between the first and the second mandibular right-molar teeth for six weeks. There were seven groups (control group, BTSc group on day 7, BTSc group on day 14, BTSc group on day 28, MSCs-BTSc group on day 7, MSCs-BTSc group on day 14, MSCs-BTSc group on day 28). The mandibular alveolar bone was analyzed and visualized in 3D with µ-CT to observe any new bone growth. Statistical Analysis. Group data were subjected to the Kruskal Wallis test followed by the Mann-Whitney (p <0.05). The µ-CT qualitative analysis shows a fibrous structure, which indicates the existence of new bone regeneration. Quantitative analysis of the periodontitis model showed a significant difference between the control model and the model with the alveolar bone resorption (p <0.05). The bone volume and density measurements revealed that the MSCs-BTSc group on day 28 formed new bone compared to other groups (p <0.05). Administration of MSCs-BTSc combination has the potential to form new alveolar bone.
Collapse
|
34
|
Panpradit N, Nilmoje T, Kasetsuwan J, Sangkhamanee SS, Surarit R. Effect of Nicotine and Porphyromonas gingivalis on the Differentiation Properties of Periodontal Ligament Fibroblasts. Eur J Dent 2021; 15:727-732. [PMID: 34331301 PMCID: PMC8630977 DOI: 10.1055/s-0041-1729678] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Objectives
This study aimed to evaluate the effect of
Porphyromonas gingivalis
and nicotine on the
in vitro
osteogenic differentiation of periodontal ligament (PDL) fibroblasts.
Materials and Methods
PDLs were cultured in Dulbecco’s modified Eagle’s medium containing 10% fetal bovine serum at 37°C under 5% CO
2
and 100% humidified atmosphere. Cells were incubated with various concentrations of nicotine and
P. gingivalis
extracts, and cell viability was determined by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide assay. To study cell differentiation, PDLs (5 × 10
4
cells) were treated with the osteogenic differentiation medium containing 10 mM β-glycerophosphate, 10 nM dexamethasone, 50 mg/mL ascorbic acid, 1 μM nicotine, and 50 µg/mL
P. gingivalis
lysate. mRNA samples were collected at 0, 7, and 14 days. Odontogenic-related gene expression, namely, Runt-related transcription factor 2 (
Runx2
), collagen type I (
COL1A1
), and alkaline phosphatase (
ALP
) was determined by reverse transcription quantitative polymerase chain reaction (RT-qPCR). Calcified nodule formation was determined on day 28 using Alizarin Red S. Analysis of variance and Tukey’s test were used to compare the difference among groups at significant level of
p
< 0.05.
Results
It showed that 50 µg/mL of
P. gingivalis
lysate and 1 µM of nicotine showed no toxicity to PDLs.
Runx2
,
COL1A1
, and
ALP
expression were found to decrease significantly after 7 days of treatment, while osteocalcin expression was found to decrease after 14 days. The nodule formation in the control group was much greater in both number and size of nodules than in experimental groups, which implied a positive sign of calcium deposition in controls.
Conclusion
The results indicated that nicotine and
P. gingivalis
showed adverse effect on osteogenic differentiation properties of PDLs.
Collapse
Affiliation(s)
- Naruemon Panpradit
- Department of Oral Medicine and Periodontology, Faculty of Dentistry, Mahidol University, Bangkok, Thailand
| | - Thanapoj Nilmoje
- Department of Oral Medicine and Periodontology, Faculty of Dentistry, Mahidol University, Bangkok, Thailand
| | - Julalux Kasetsuwan
- Department of Oral Medicine and Periodontology, Faculty of Dentistry, Mahidol University, Bangkok, Thailand.,Section of Periodontology, College of Dental Medicine, Rangsit University, Patumthani, Thailand
| | | | - Rudee Surarit
- Department of Oral Biology, Faculty of Dentistry, Mahidol University, Bangkok, Thailand
| |
Collapse
|
35
|
Milewska A, Sigurjonsson OE, Leosson K. SERS Imaging of Mesenchymal Stromal Cell Differentiation. ACS APPLIED BIO MATERIALS 2021; 4:4999-5007. [PMID: 35007048 DOI: 10.1021/acsabm.1c00286] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Understanding the process of mesenchymal stromal cell (MSC) osteogenic differentiation is essential for a wide range of medical applications. However, these primary cells vary significantly from donor to donor, making it difficult to fully exploit their therapeutic potential. Although osteogenic differentiation has been studied extensively, there is still a shortage of standardized methods for the evaluation of the degree of differentiation. Here, we employ noninvasive surface-enhanced Raman scattering (SERS) for studying such cells, offering a better understanding of cellular processes in situ. We present the long-term differentiation of MSCs on biocompatible gold nanoisland SERS substrates, combining imaging of cells with spectroscopic detection of molecular species and chemical events occurring on the cellular membrane adjacent to the surface of the SERS substrate. We detect multiple signs of bone tissue formation, from an early stage to mature osteoblasts, without labeling. We show that the results correlate very well with classical differentiation-detecting assays, indicating that the SERS imaging technique alone is sufficient to study the progress of osteogenic differentiation of such cells, paving a way toward continuous label-free screening of live cells.
Collapse
Affiliation(s)
- Adrianna Milewska
- Innovation Center Iceland, Árleynir 2-8, 112 Reykjavík, Iceland.,The Blood Bank, Landspitali University Hospital, Snorrabraut 60, 105 Reykjavík, Iceland.,University of Iceland, School of Engineering and Natural Sciences, Sæmundargötu 2, 101 Reykjavík, Iceland
| | - Olafur E Sigurjonsson
- The Blood Bank, Landspitali University Hospital, Snorrabraut 60, 105 Reykjavík, Iceland.,Reykjavik University, School of Science and Engineering, Menntavegur 1, 101 Reykjavík, Iceland
| | | |
Collapse
|
36
|
In Vivo and In Vitro Analyses of Titanium-Hydroxyapatite Functionally Graded Material for Dental Implants. BIOMED RESEARCH INTERNATIONAL 2021; 2021:8859945. [PMID: 34036104 PMCID: PMC8121567 DOI: 10.1155/2021/8859945] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Revised: 02/21/2021] [Accepted: 03/17/2021] [Indexed: 11/18/2022]
Abstract
Purpose The stress shielding effect caused due to the mechanical mismatch between the solid titanium and the surrounding bone tissue warrants the utilization of a mechanically and biologically compatible material such as the titanium-hydroxyapatite (Ti-HA) functionally graded material (FGM) for dental implants. This study is aimed at fabricating a Ti-HA FGM with superior mechanical and biological properties for dental implantation. Materials and Methods We fabricated a Ti-HA FGM with different Ti volume fractions (VFs) using HA and Ti powders. Ti-HA was characterized by studying its mechanical properties. Cytotoxicity was examined using a Cell Counting Kit-8 assay and an LDH cell cytotoxicity assay. Scanning electron microscopy was performed on an XL30 environmental scanning electron microscope (ESEM). Alkaline phosphatase (ALP) and transforming growth factor (TGF-β1) expressions were quantitatively monitored using enzyme-linked immunosorbent assay (ELISA) kits. The expressions of TGF-β receptors and ALP genes were measured using real-time polymerase chain reaction. The Ti-HA FGM dental implants were placed in beagle dogs. Microcomputed tomography (CT) and hard tissue slices were performed to evaluate the bone-implant contact (BIC) and bone volume over total volume (BV/TV). Results The density and mechanical properties of the Ti-HA exhibited various graded distributions corresponding to VF. Based on the results of the Cell Counting Kit-8 (CCK-8) and lactate dehydrogenase (LDH) assays, the difference in cytotoxicity between the two groups was statistically nonsignificant (P = 0.11). The ALP and TGF-β1 levels were slightly upregulated. The transcript levels of ALP and TGF-βRI were higher in the Ti-HA groups than in the Ti group at 7 days, whereas the transcript levels of TGF-βRII exhibited no obvious increase. The BIC did not exhibit significant differences between the Ti and Ti-HA FGM groups (P = 0.0504). BV/TV showed the Ti-HA FGM group had better osteogenesis (P = 0.04). Conclusion Ti-HA FGM contributes to the osteogenesis of dental implants in vivo and in vitro.
Collapse
|
37
|
Efficacy of chitinase-3-like protein 1 as an in vivo bone formation predictable marker of maxillary/mandibular bone marrow stromal cells. Regen Ther 2021; 18:38-50. [PMID: 33869686 PMCID: PMC8027134 DOI: 10.1016/j.reth.2021.03.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 03/01/2021] [Accepted: 03/10/2021] [Indexed: 12/11/2022] Open
Abstract
Introduction Maxillary/mandibular bone marrow stromal cells (MBMSCs) are a useful cell source for bone regeneration in the oral and maxillofacial region. To further ensure the clinical application of MBMSCs in bone regenerative therapy, it is important to determine the bone formation capacity of MBMSCs before transplantation. The aim of this study is to identify the molecular marker that determines the in vivo bone formation capacity of MBMSCs. Methods The cell growth, cell surface antigens, in vitro and in vivo bone formation capacity of MBMSCs were examined. The amount of chitinase-3-like protein 1 (CHI3L1) secreted into the conditioned medium was quantified. The effects of CHI3L1 on the cell growth and osteogenic differentiation potential of MBMSCs and on the cell growth and migration of vascular endothelial cells and fibroblasts were examined. Results The cell growth, and in vitro and in vivo bone formation capacity of the cells treated with different conditions were observed. MBMSCs that secreted a large amount of CHI3L1 into the conditioned medium tended to have low in vivo bone formation capacity, whereas MBMSCs that secreted a small amount of CHI3L1 had greater in vivo bone formation capacity. CHI3L1 promoted the migration of vascular endothelial cells, and the cell growth and migration of fibroblasts. Conclusion Our study indicates that the in vitro osteogenic differentiation capacity of MBMSCs and the in vivo bone formation capacities of MBMSCs were not necessarily correlated. The transplantation of high CHI3L1 secretory MBMSCs may suppress bone formation by inducing fibrosis at the site. These results suggest that the CHI3L1 secretion levels from MBMSCs may be used as a predictable marker of bone formation capacity in vivo. In vitro and in vivo bone formation capacities of MBMSCs were not correlated. MBMSCs with high CHI3L1 secretion tended to have low in vivo bone formation. MBMSCs with low CHI3L1 secretion tended to have high in vivo bone formation. CHI3L1 can be in vivo bone formation capacity predictable marker of MBMSCs.
Collapse
Key Words
- ALP, Alkaline phosphatase
- BMSC, bone marrow-derived stem cell
- Bone formation capacity
- CHI3L1, chitinase-3-like protein 1
- Chitinase-3-like protein 1
- FBS, fetal bovine serum
- HUVEC, human umbilical vein endothelial cells
- Jaw bone marrow stromal cells
- MBMSC, maxillary/mandibular bone marrow stromal cells
- MSCs, mesenchymal stem cells
- Migration
- NHDF, normal human dermal fibroblasts
- α-MEM, alpha modified Eagle's minimum essential medium
- β-TCP, beta-tricalcium phosphate
Collapse
|
38
|
Khokhani P, Rahmani NR, Kok A, Öner FC, Alblas J, Weinans H, Kruyt MC, Croes M. Use of Therapeutic Pathogen Recognition Receptor Ligands for Osteo-Immunomodulation. MATERIALS 2021; 14:ma14051119. [PMID: 33673651 PMCID: PMC7957819 DOI: 10.3390/ma14051119] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 02/22/2021] [Indexed: 01/08/2023]
Abstract
Therapeutic pathogen recognition receptor (PRR) ligands are reaching clinical practice following their ability to skew the immune response in a specific direction. We investigated the effects of various therapeutic PRR ligands on bone cell differentiation and inflammation. Following stimulation, alkaline phosphatase (ALP) activity (Day 10), osteocalcin, osteonectin expression (Day 14), and calcium deposition (Day 21) were quantified in bone marrow-derived human mesenchymal stem cells (hMSCs). The osteoclastogenic response was determined by measuring tartrate-resistant acid phosphate (TRAP) activity in human monocytes. TNF-α, IL-6, IL-8, and IL-10 expressions were measured by enzyme-linked immunosorbent assay as an indicator of the ligands’ inflammatory properties. We found that nucleic acid-based ligands Poly(I:C) and CpG ODN C increased early ALP activity in hMSCs by 4-fold without affecting osteoclast formation. These ligands did not enhance expression of the other, late osteogenic markers. MPLA, Curdlan, and Pam3CSK4 did not affect osteogenic differentiation, but inhibited TRAP activity in monocytes, which was associated with increased expression of all measured cytokines. Nucleic acid-based ligands are identified as the most promising osteo-immunomodulators, as they favor early osteogenic differentiation without inducing an exaggerated immune-cell mediated response or interfering in osteoclastogenesis and thus can be potentially harnessed for multifunctional coatings for bone biomaterials.
Collapse
Affiliation(s)
- Paree Khokhani
- Department of Orthopedics, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| | - Nada R Rahmani
- Department of Orthopedics, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| | - Anne Kok
- Department of Orthopedics, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| | - F Cumhur Öner
- Department of Orthopedics, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| | - Jacqueline Alblas
- Department of Orthopedics, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| | - Harrie Weinans
- Department of Orthopedics, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
- Department of Biomechanical Engineering, Technical University Delft, 2628 CD Delft, The Netherlands
| | - Moyo C Kruyt
- Department of Orthopedics, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| | - Michiel Croes
- Department of Orthopedics, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| |
Collapse
|
39
|
Al-Maawi S, Dohle E, Lim J, Weigl P, Teoh SH, Sader R, Ghanaati S. Biologization of Pcl-Mesh Using Platelet Rich Fibrin (Prf) Enhances Its Regenerative Potential In Vitro. Int J Mol Sci 2021; 22:2159. [PMID: 33671550 PMCID: PMC7926906 DOI: 10.3390/ijms22042159] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 02/16/2021] [Accepted: 02/18/2021] [Indexed: 11/17/2022] Open
Abstract
INTRODUCTION Resorbable synthetic scaffolds are promising for different indications, especially in the context of bone regeneration. However, they require additional biological components to enhance their osteogenic potential. In addition to different cell types, autologous blood-derived matrices offer many advantages to enhance the regenerative capacity of biomaterials. The present study aimed to analyze whether biologization of a PCL-mesh coated using differently centrifuged Platelet rich fibrin (PRF) matrices will have a positive influence on primary human osteoblasts activity in vitro. A polymeric resorbable scaffold (Osteomesh, OsteoporeTM (OP), Singapore) was combined with differently centrifuged PRF matrices to evaluate the additional influence of this biologization concept on bone regeneration in vitro. Peripheral blood of three healthy donors was used to gain PRF matrices centrifuged either at High (710× g, 8 min) or Low (44× g, 8 min) relative centrifugal force (RCF) according to the low speed centrifugation concept (LSCC). OP-PRF constructs were cultured with pOBs. POBs cultured on the uncoated OP served as a control. After three and seven days of cultivation, cell culture supernatants were collected to analyze the pOBs activity by determining the concentrations of VEGF, TGF-β1, PDGF, OPG, IL-8, and ALP- activity. Immunofluorescence staining was used to evaluate the Osteopontin expression of pOBs. After three days, the group of OP+PRFLow+pOBs showed significantly higher expression of IL-8, TGF-ß1, PDGF, and VEGF compared to the group of OP+PRFHigh+pOBs and OP+pOBs. Similar results were observed on day 7. Moreover, OP+PRFLow+pOBs exhibited significantly higher activity of ALP compared to OP+PRFHigh+pOBs and OP+pOBs. Immunofluorescence staining showed a higher number of pOBs adherent to OP+PRFLow+pOBs compared to the groups OP+PRFHigh+pOBs and OP+pOBs. To the best of our knowledge, this study is the first to investigate the osteoblasts activity when cultured on a PRF-coated PCL-mesh in vitro. The presented results suggest that PRFLow centrifuged according to LSCC exhibits autologous blood cells and growth factors, seem to have a significant effect on osteogenesis. Thereby, the combination of OP with PRFLow showed promising results to support bone regeneration. Further in vivo studies are required to verify the results and carry out potential results for clinical translation.
Collapse
Affiliation(s)
- Sarah Al-Maawi
- FORM, Frankfurt Oral Regenerative Medicine, Clinic for Maxillofacial and Plastic Surgery, Goethe University, Theodor-Stern-Kai 7, 60596 Frankfurt, Germany; (S.A.-M.); (E.D.); (R.S.)
| | - Eva Dohle
- FORM, Frankfurt Oral Regenerative Medicine, Clinic for Maxillofacial and Plastic Surgery, Goethe University, Theodor-Stern-Kai 7, 60596 Frankfurt, Germany; (S.A.-M.); (E.D.); (R.S.)
| | - Jing Lim
- Osteopore International, Singapore 618305, Singapore;
| | - Paul Weigl
- Department of Prosthodontics and Head of Department of Postgraduate Education, Center for Dentistry and Oral Medicine (Carolinum), Goethe University, Theodor-Stern-Kai 7, 60596 Frankfurt, Germany;
| | - Swee Hin Teoh
- School of Chemical and Biomedical Engineering/Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 637459, Singapore;
| | - Robert Sader
- FORM, Frankfurt Oral Regenerative Medicine, Clinic for Maxillofacial and Plastic Surgery, Goethe University, Theodor-Stern-Kai 7, 60596 Frankfurt, Germany; (S.A.-M.); (E.D.); (R.S.)
| | - Shahram Ghanaati
- FORM, Frankfurt Oral Regenerative Medicine, Clinic for Maxillofacial and Plastic Surgery, Goethe University, Theodor-Stern-Kai 7, 60596 Frankfurt, Germany; (S.A.-M.); (E.D.); (R.S.)
| |
Collapse
|
40
|
Mohammadi B, Esmaeilizade Z, Omrani MD, Ghaderian SMH, Rajabibazl M, Fazeli Z. The Effect of Co-treating Human Mesenchymal Stem Cells with Epigallocatechin Gallate and Hypoxia-Inducible Factor-1 on the Expression of RANKL/RANK/OPG Signaling Pathway, Osteogenesis, and Angiogenesis Genes. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2021. [DOI: 10.1007/s40883-021-00197-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
41
|
Palekar G, Bhalodiya HP, Archik S, Trivedi K. Retrospective Study on Implantation of Autologous-Cultured Osteoblasts for the Treatment of Patients with Avascular Necrosis of the Femoral Head. Orthop Res Rev 2021; 13:15-23. [PMID: 33568953 PMCID: PMC7868257 DOI: 10.2147/orr.s281030] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 12/11/2020] [Indexed: 11/24/2022] Open
Abstract
Purpose Osteonecrosis of the femoral head is a progressive and debilitating disease that causes pain, osteoarthritis and hip joint collapse, eventually necessitating hip replacement. This study evaluated the long-term outcomes of autologous adult live-cultured osteoblasts (AALCO) implantation in patients with osteonecrosis of femoral head. Patients and Methods In this retrospective multicenter study, we collated and analyzed data of patients ≥12 years of age who underwent AALCO (OSSGROW®) between 2010 and 2015 for the treatment of osteonecrosis of the femoral head. Results Data from 64 patients (101 hip joints) were assessed in this study. The mean ±SD duration of disease since diagnosis of osteonecrosis was 7.4±1.6 years. The mean follow-up duration was 6.3±1.4 years. The mean VAS score (n=98 hips) reduced significantly from 58.8 ± 13.8 to 32.2 ± 32.1 post-operatively (mean difference: −26.5±35.2, p=0.001) and Harris hip score (n=97 hips) also significantly improved from 47.1±12.3 to 63.7±27.7 post-operatively (mean difference:16.7±28.7, p=0.0001). Following the AALCO treatment, 29 hips in 13 patients (28.7%) underwent total hip replacement (THA), indicating that AALCO treatment could delay THA for 71.3% of hips. A total of 39.1% of hips diagnosed in early stage versus 60.8% in the late stage of osteonecrosis required THA. Overall, 60.4% of hips improved, 2% remained stable, and 37.6% progressed following the AALCO implantation. The condition of disease was found to have improved in 71.1% of patients in early stage (Grades I and II) versus 58% in the late stage (Grades III and IV) of osteonecrosis. Conclusion Patients with osteonecrosis who received implantation of autologous-cultured osteoblasts using AALCO showed improvement in joint function and decrease in pain. Treatment with AALCO halted progression of osteonecrosis, preserved the natural hip, and eliminated the need for hip replacement surgeries in 7 out of 10 patients.
Collapse
Affiliation(s)
- Gauresh Palekar
- Orthopedic Surgeon, Surya Orthopedic Clinic, Mumbai, Maharashtra, India
| | - H P Bhalodiya
- Orthopedic Surgeon, Walk-Fit Saviour Hospital, Ahmedabad, Gujarat, India
| | - Shreedhar Archik
- Orthopedic Surgeon, Dr. Sreedhar Aarchik Orthopedic Care Clinic, Dadar West, Mumbai, Maharashtra, India
| | - Kalpesh Trivedi
- Arthroscopy Surgeon, Aastha Arthroscopy and Joint Surgery Clinic, Ahmedabad, Gujarat, India
| |
Collapse
|
42
|
Omar AE, Ibrahim AM, Abd El-Aziz TH, Al-Rashidy ZM, Farag MM. Role of alkali metal oxide type on the degradation and in vivo biocompatibility of soda-lime-borate bioactive glass. J Biomed Mater Res B Appl Biomater 2020; 109:1059-1073. [PMID: 33274827 DOI: 10.1002/jbm.b.34769] [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: 07/07/2020] [Revised: 10/24/2020] [Accepted: 11/17/2020] [Indexed: 11/09/2022]
Abstract
In this work, it is the first time to study the effect of replacing of Na2 O by a fixed amount of Li2 O or K2 O in soda-lime-borate glass on its in vivo biocompatibility. The glass composition was based on xM2 O-20x Na2 O20 CaO60 B2 O3 , (wt %), where, M2 OLi2 O and K2 O, and consequently, samples encoded BN100, BK50, and BL50. The degradation test was carried out in 0.25 M K2 HPO4 solution. The in vivo test was performed in the femoral bone defect of Sprague-Dawley adult male rat. Following up bone formation was conducted by the histological analyses and bone formation markers (alkaline phosphatase [ALP] and osteocalcin [OCN]). Furthermore, the glass effect on the liver and kidney functions was addressed in this study using (alanine transaminase [ALT] and aspartate transaminase [AST]) and (urea and creatinine), respectively. The results of the degradation test showed that the glass dissolution rate was increased by incorporating of K2 O, and its ion release was occurred by a diffusion-controlled process. Moreover, in vivo bioactivity test showed that serum activity of ALP, OCN level, and the newly formed bone was higher in BL50-implanted group than that of BN100 andBK50at 3 w and 6 w post-surgery. As well as, implantation of all glass samples in the femoral bone defect did not alter the liver and kidney functions. In conclusion, the synthesized borate glass was well served as a controlled delivery system for Li+ ion release, which enhanced bone formation as shown from the bone formation markers (ALP and OCN).
Collapse
Affiliation(s)
- Areg E Omar
- Department of Physics, Faculty of Science, Al-Azhar University (Girls' Branch), Nasr City, Egypt
| | - Ahlam M Ibrahim
- Physics Department (Biophysics Branch), Faculty of Science, Al-Azhar University (Girls' Branch), Nasr City, Egypt
| | - Tamer H Abd El-Aziz
- Department of Parasitology and Animal Diseases, Veterinary Research Division, National Research Centre, Giza, Egypt
| | - Zainab M Al-Rashidy
- Department of Refractoriness, Ceramics and Building Materials, National Research Centre, Giza, Egypt
| | - Mohammad M Farag
- Glass Research Department, National Research Centre, Giza, Egypt
| |
Collapse
|
43
|
miR-124-3p promotes BMSC osteogenesis via suppressing the GSK-3β/β-catenin signaling pathway in diabetic osteoporosis rats. In Vitro Cell Dev Biol Anim 2020; 56:723-734. [PMID: 33085064 DOI: 10.1007/s11626-020-00502-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 08/31/2020] [Indexed: 10/23/2022]
Abstract
The purpose of this study is to investigate miRNAs' effects, targeting the Wnt signaling pathway, on osteogenic differentiation to provide new targets for diabetic osteoporosis treatments. Twelve male rats were divided into a normal rat group (NOR group) and a model rat group (MOD group). Cluster analysis of differentially expressed miRNAs and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis were performed. Primary rat bone marrow mesenchymal stem cells (BMSCs) were divided into a high-glucose group and a low-glucose group, and osteogenic differentiation was induced. Alkaline phosphatase (ALP) staining and Alizarin Red staining were used for pathological analysis of the cells. Western blot analysis was used to measure GSK-3β, β-catenin, p-β-catenin, c-Myc, and CyclinD1 expression. Immunofluorescence (IF) was used to analyze the effect of GSK-3β inhibitor (CHIR99021) on β-catenin and CyclinD1 expressions levels in BMSCs. A total of 428 differentially expressed miRNAs were found between the NOR and MOD groups. KEGG analysis showed that the target genes were mostly enriched in signaling pathways, including PI3K-Akt, focal adhesion, AGE-RAGE, HIF-1, and Wnt. qPCR verification demonstrated that miR-124-3p exhibited the greatest difference in expression level. In BMSCs, miR-124-3p overexpression could reverse the inhibited expression of BMSC osteogenic markers, including Alpl, Bglap, and Runx2, induced by high glucose. Western blot analysis revealed that the transfection of miR-124-3p mimics could further reverse the upregulated p-β-catenin and GSK-3β levels and the downregulated c-Myc and CyclinD1 levels induced by high glucose. IF results revealed that BMSCs treated CHIR99021 under high glucose showed the reduced GSK-3β and increased β-catenin and CyclinD1 expression levels. Our research highlighted miRNAs' important roles in regulating the Wnt pathway and provided new information for the diagnosis and treatment of diabetic osteoporosis.
Collapse
|
44
|
Assessment of the Release Profile of Fibroblast Growth Factor-2-Load Mesoporous Calcium Silicate/Poly-ε-caprolactone 3D Scaffold for Regulate Bone Regeneration. Processes (Basel) 2020. [DOI: 10.3390/pr8101249] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Recent advances in three-dimensional printing technology enable facile and on-demand fabrication of patient-specific bone scaffolds. However, there is still an urgent need for printable biomaterials with osteoinductivity. In the present study, we propose an approach to synthesize fibroblast growth factor-2 loaded-mesoporous calcium silicate nanoparticles. The growth factor loaded-nanoparticles served as fillers of polycaprolactone and then the composite scaffolds with a controlled pore structure were obtained through a fused deposition modeling technique. To evaluate the feasibility of the composite scaffolds in bone tissue engineering, drug release kinetic, bioactivity, cell proliferation, differentiation, and animal study were conducted. Our findings illustrate that utilization of mesoporous calcium silicate allowed the introduction of fibroblast growth factor-2 into the composite scaffolds through a simple soaking process and then gradually released from the scaffold to facilitate proliferation and osteogenesis differentiation of human Wharton’s jelly mesenchymal stem cells. Additionally, the in vivo femur defect experiments also indicate that the co-existence of calcium silicate and fibrous growth factor-2 synergistically accelerated new bone formation. These results demonstrate that the fibroblast growth factor-2-loaded mesoporous calcium silicate nanoparticles/polycaprolactone composite scaffolds may serve as potential bone grafts for facilitating repair of defected bone tissues.
Collapse
|
45
|
Al-Rashidy ZM, Omar AE, El-Aziz THA, Farag MM. In vivo bioactivity assessment of strontium-containing soda-lime-borate glass implanted in femoral defect of rat. J Inorg Organomet Polym Mater 2020. [DOI: 10.1007/s10904-020-01535-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
46
|
Osteoblastic exosomes. A non-destructive quantitative approach of alkaline phosphatase to assess osteoconductive nanomaterials. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 115:110931. [DOI: 10.1016/j.msec.2020.110931] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Revised: 03/02/2020] [Accepted: 04/03/2020] [Indexed: 11/18/2022]
|
47
|
A multifaceted biomimetic interface to improve the longevity of orthopedic implants. Acta Biomater 2020; 110:266-279. [PMID: 32344174 DOI: 10.1016/j.actbio.2020.04.020] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 03/23/2020] [Accepted: 04/09/2020] [Indexed: 01/22/2023]
Abstract
The rise of additive manufacturing has provided a paradigm shift in the fabrication of precise, patient-specific implants that replicate the physical properties of native bone. However, eliciting an optimal biological response from such materials for rapid bone integration remains a challenge. Here we propose for the first time a one-step ion-assisted plasma polymerization process to create bio-functional 3D printed titanium (Ti) implants that offer rapid bone integration. Using selective laser melting, porous Ti implants with enhanced bone-mimicking mechanical properties were fabricated. The implants were functionalized uniformly with a highly reactive, radical-rich polymeric coating generated using a unique combination of plasma polymerization and plasma immersion ion implantation. We demonstrated the performance of such activated Ti implants with a focus on the coating's homogeneity, stability, and biological functionality. It was shown that the optimized coating was highly robust and possessed superb physico-chemical stability in a corrosive physiological solution. The plasma activated coating was cytocompatible and non-immunogenic; and through its high reactivity, it allowed for easy, one-step covalent immobilization of functional biomolecules in the absence of solvents or chemicals. The activated Ti implants bio-functionalized with bone morphogenetic protein 2 (BMP-2) showed a reduced protein desorption and a more sustained osteoblast response both in vitro and in vivo compared to implants modified through conventional physisorption of BMP-2. The versatile new approach presented here will enable the development of bio-functionalized additively manufactured implants that are patient-specific and offer improved integration with host tissue. STATEMENT OF SIGNIFICANCE: Additive manufacturing has revolutionized the fabrication of patient-specific orthopedic implants. Although such 3D printed implants can show desirable mechanical and mass transport properties, they often require surface bio-functionalities to enable control over the biological response. Surface covalent immobilization of bioactive molecules is a viable approach to achieve this. Here we report the development of additively manufactured titanium implants that precisely replicate the physical properties of native bone and are bio-functionalized in a simple, reagent-free step. Our results show that covalent attachment of bone-related growth factors through ion-assisted plasma polymerized interlayers circumvents their desorption in physiological solution and significantly improves the bone induction by the implants both in vitro and in vivo.
Collapse
|
48
|
Avery SJ, Ayre WN, Sloan AJ, Waddington RJ. Interrogating the Osteogenic Potential of Implant SurfacesIn Vitro: A Review of Current Assays. TISSUE ENGINEERING PART B-REVIEWS 2020; 26:217-229. [DOI: 10.1089/ten.teb.2019.0312] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Steven James Avery
- Department of Oral and Biomedical Sciences, College of Biomedical and Life Sciences, Cardiff Institute for Tissue Engineering and Repair, School of Dentistry, Cardiff University, Cardiff, United Kingdom
| | - Wayne Nishio Ayre
- Department of Oral and Biomedical Sciences, College of Biomedical and Life Sciences, Cardiff Institute for Tissue Engineering and Repair, School of Dentistry, Cardiff University, Cardiff, United Kingdom
| | - Alastair James Sloan
- Department of Oral and Biomedical Sciences, College of Biomedical and Life Sciences, Cardiff Institute for Tissue Engineering and Repair, School of Dentistry, Cardiff University, Cardiff, United Kingdom
| | - Rachel Jane Waddington
- Department of Oral and Biomedical Sciences, College of Biomedical and Life Sciences, Cardiff Institute for Tissue Engineering and Repair, School of Dentistry, Cardiff University, Cardiff, United Kingdom
| |
Collapse
|
49
|
Enhancing Activity of Pleurotus sajor-caju (Fr.) Sing β-1,3-Glucanoligosaccharide ( Ps-GOS) on Proliferation, Differentiation, and Mineralization of MC3T3-E1 Cells through the Involvement of BMP-2/Runx2/MAPK/Wnt/β-Catenin Signaling Pathway. Biomolecules 2020; 10:biom10020190. [PMID: 32012654 PMCID: PMC7072289 DOI: 10.3390/biom10020190] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 01/24/2020] [Accepted: 01/24/2020] [Indexed: 12/21/2022] Open
Abstract
Osteoporosis is a leading world health problem that results from an imbalance between bone formation and bone resorption. β-glucans has been extensively reported to exhibit a wide range of biological activities, including antiosteoporosis both in vitro and in vivo. However, the molecular mechanisms responsible for β-glucan-mediated bone formation in osteoblasts have not yet been investigated. The oyster mushroom Pleurotus sajor-caju produces abundant amounts of an insoluble β-glucan, which is rendered soluble by enzymatic degradation using Hevea glucanase to generate low-molecular-weight glucanoligosaccharide (Ps-GOS). This study aimed to investigate the osteogenic enhancing activity and underlining molecular mechanism of Ps-GOS on osteoblastogenesis of pre-osteoblastic MC3T3-E1 cells. In this study, it was demonstrated for the first time that low concentrations of Ps-GOS could promote cell proliferation and division after 48 h of treatment. In addition, Ps-GOS upregulated the mRNA and protein expression level of bone morphogenetic protein-2 (BMP-2) and runt-related transcription factor-2 (Runx2), which are both involved in BMP signaling pathway, accompanied by increased alkaline phosphatase (ALP) activity and mineralization. Ps-GOS also upregulated the expression of osteogenesis related genes including ALP, collagen type 1 (COL1), and osteocalcin (OCN). Moreover, our novel findings suggest that Ps-GOS may exert its effects through the mitogen-activated protein kinase (MAPK) and wingless-type MMTV integration site (Wnt)/β-catenin signaling pathways.
Collapse
|
50
|
Huang X, Chen M, Wu H, Jiao Y, Zhou C. Macrophage Polarization Mediated by Chitooligosaccharide (COS) and Associated Osteogenic and Angiogenic Activities. ACS Biomater Sci Eng 2020; 6:1614-1629. [DOI: 10.1021/acsbiomaterials.9b01550] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Xiuhong Huang
- Department of Materials Science and Engineering, Jinan University, Guangzhou 510632, China
| | - Meng Chen
- Department of Materials Science and Engineering, Jinan University, Guangzhou 510632, China
| | - Haoming Wu
- Department of Materials Science and Engineering, Jinan University, Guangzhou 510632, China
| | - Yanpeng Jiao
- Department of Materials Science and Engineering, Jinan University, Guangzhou 510632, China
| | - Changren Zhou
- Department of Materials Science and Engineering, Jinan University, Guangzhou 510632, China
| |
Collapse
|