1
|
Sharma P, Mishra V, Murab S. Unlocking Osseointegration: Surface Engineering Strategies for Enhanced Dental Implant Integration. ACS Biomater Sci Eng 2025; 11:67-94. [PMID: 39620938 DOI: 10.1021/acsbiomaterials.4c01178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2025]
Abstract
Tooth loss is a prevalent problem faced by individuals of all ages across the globe. Various biomaterials, such as metals, bioceramics, polymers, composites of ceramics and polymers, etc., have been used for the manufacturing of dental implants. The success of a dental implant primarily depends on its osseointegration rate. The current surface modification techniques fail to imbibe the basics of tooth development, which can impart better mineralization and osseointegration. This can be improved by developing an understanding of the developmental pathways of dental tissue. Stimulating the correct signaling pathways through inductive material systems can bring about a paradigm shift in dental implant materials. The current review focuses on the developmental pathway and mineralization process that happen during tooth formation and how surface modifications can help in biomimetic mineralization, thereby enhancing osseointegration. We further describe the effect of dental implant surface modifications on mineralization, osteoinduction, and osseointegration; both in vitro and in vivo. The review will help us to understand the natural process of teeth development and mineralization and how the surface properties of dental implants can be further improved to mimic teeth development, in turn increasing osseointegration.
Collapse
Affiliation(s)
- Pankaj Sharma
- School of Biosciences & Bioengineering, Indian Institute of Technology Mandi, Kamand HP-175075, India
| | - Vedante Mishra
- School of Biosciences & Bioengineering, Indian Institute of Technology Mandi, Kamand HP-175075, India
| | - Sumit Murab
- School of Biosciences & Bioengineering, Indian Institute of Technology Mandi, Kamand HP-175075, India
- Indian Knowledge System and Mental Health Applications Centre, Indian Institute of Technology Mandi, Kamand HP-175075, India
- BioX Centre, Indian Institute of Technology Mandi, Kamand HP-175075, India
- Advanced Materials Research Centre, Indian Institute of Technology Mandi, Kamand HP-175075, India
- Technology Innovation Hub in Human-Computer Interaction (iHub), Kamand HP-175075, India
| |
Collapse
|
2
|
Pandit A, Indurkar A, Locs J, Haugen HJ, Loca D. Calcium Phosphates: A Key to Next-Generation In Vitro Bone Modeling. Adv Healthc Mater 2024; 13:e2401307. [PMID: 39175382 PMCID: PMC11582516 DOI: 10.1002/adhm.202401307] [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: 04/09/2024] [Revised: 08/06/2024] [Indexed: 08/24/2024]
Abstract
The replication of bone physiology under laboratory conditions is a prime target behind the development of in vitro bone models. The model should be robust enough to elicit an unbiased response when stimulated experimentally, giving reproducible outcomes. In vitro bone tissue generation majorly requires the availability of cellular components, the presence of factors promoting cellular proliferation and differentiation, efficient nutrient supply, and a supporting matrix for the cells to anchor - gaining predefined topology. Calcium phosphates (CaP) are difficult to ignore while considering the above requirements of a bone model. Therefore, the current review focuses on the role of CaP in developing an in vitro bone model addressing the prerequisites of bone tissue generation. Special emphasis is given to the physico-chemical properties of CaP that promote osteogenesis, angiogenesis and provide sufficient mechanical strength for load-bearing applications. Finally, the future course of action is discussed to ensure efficient utilization of CaP in the in vitro bone model development field.
Collapse
Affiliation(s)
- Ashish Pandit
- Institute of Biomaterials and BioengineeringFaculty of Natural Sciences and TechnologyRiga Technical UniversityPulka Street 3RigaLV‐1007Latvia
- Baltic Biomaterials Centre of ExcellenceHeadquarters at Riga Technical UniversityRigaLV‐1007Latvia
| | - Abhishek Indurkar
- Institute of Biomaterials and BioengineeringFaculty of Natural Sciences and TechnologyRiga Technical UniversityPulka Street 3RigaLV‐1007Latvia
- Baltic Biomaterials Centre of ExcellenceHeadquarters at Riga Technical UniversityRigaLV‐1007Latvia
| | - Janis Locs
- Institute of Biomaterials and BioengineeringFaculty of Natural Sciences and TechnologyRiga Technical UniversityPulka Street 3RigaLV‐1007Latvia
- Baltic Biomaterials Centre of ExcellenceHeadquarters at Riga Technical UniversityRigaLV‐1007Latvia
| | | | - Dagnija Loca
- Institute of Biomaterials and BioengineeringFaculty of Natural Sciences and TechnologyRiga Technical UniversityPulka Street 3RigaLV‐1007Latvia
- Baltic Biomaterials Centre of ExcellenceHeadquarters at Riga Technical UniversityRigaLV‐1007Latvia
| |
Collapse
|
3
|
Mishra A, Kumar R, Harilal S, Nigam M, Datta D, Singh S. Emerging Landscape of In Vitro Models for Assessing Rheumatoid Arthritis Management. ACS Pharmacol Transl Sci 2024; 7:2280-2305. [PMID: 39144547 PMCID: PMC11320735 DOI: 10.1021/acsptsci.4c00260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 06/23/2024] [Accepted: 06/24/2024] [Indexed: 08/16/2024]
Abstract
Rheumatoid arthritis (RA) is a complex condition that is influenced by various causes, including immunological, genetic, and environmental factors. Several studies using animal models have documented immune system dysfunction and described the clinical characteristics of the disease. These studies have provided valuable insights into the pathogenesis of inflammatory arthritis and the identification of new targets for treatment. Nevertheless, none of these animal models successfully replicated all the characteristics of RA. Additionally, numerous experimental medications, which were developed based on our enhanced comprehension of the immune system's function in RA, have shown potential in animal research but ultimately proved ineffective during different stages of clinical trials. There have been several novel therapy alternatives, which do not achieve a consistently outstanding therapeutic outcome in all patients. This underscores the importance of employing the progress in in vitro models, particularly 3D models like tissue explants, and diverse multicomponent approaches such as coculture strategies, synovial membrane, articular cartilage, and subchondral bone models that accurately replicate the structural characteristics of RA pathophysiology. These methods are crucial for the advancement of potential therapeutic strategies. This review discusses the latest advancements in in vitro models and their potential to greatly impact research on managing RA.
Collapse
Affiliation(s)
- Abhay
Prakash Mishra
- Department
of Pharmacology, University of Free State, Bloemfontein 9301, South Africa
- Department
of Pharmaceutical Sciences, Naresuan University, Phitsanulok 65000, Thailand
| | - Rajesh Kumar
- Faculty
of Pharmaceutical Sciences, Kerala University
of Health Sciences, Kerala 680596, India
| | - Seetha Harilal
- Faculty
of Pharmaceutical Sciences, Kerala University
of Health Sciences, Kerala 680596, India
| | - Manisha Nigam
- Department
of Biochemistry, Hemvati Nandan Bahuguna
Garhwal University, Srinagar
Garhwal, Uttarakhand 246174, India
| | - Deepanjan Datta
- Department
of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| | - Sudarshan Singh
- Office of
Research Administration, Chiang Mai University, Chiang Mai 50200, Thailand
- Faculty of
Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand
| |
Collapse
|
4
|
Alonso-Fernández I, Haugen HJ, Nogueira LP, López-Álvarez M, González P, López-Peña M, González-Cantalapiedra A, Muñoz-Guzón F. Enhanced Bone Healing in Critical-Sized Rabbit Femoral Defects: Impact of Helical and Alternate Scaffold Architectures. Polymers (Basel) 2024; 16:1243. [PMID: 38732711 PMCID: PMC11085737 DOI: 10.3390/polym16091243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 04/20/2024] [Accepted: 04/26/2024] [Indexed: 05/13/2024] Open
Abstract
This study investigates the effect of scaffold architecture on bone regeneration, focusing on 3D-printed polylactic acid-bioceramic calcium phosphate (PLA-bioCaP) composite scaffolds in rabbit femoral condyle critical defects. We explored two distinct scaffold designs to assess their influence on bone healing and scaffold performance. Structures with alternate (0°/90°) and helical (0°/45°/90°/135°/180°) laydown patterns were manufactured with a 3D printer using a fused deposition modeling technique. The scaffolds were meticulously characterized for pore size, strut thickness, porosity, pore accessibility, and mechanical properties. The in vivo efficacy of these scaffolds was evaluated using a femoral condyle critical defect model in eight skeletally mature New Zealand White rabbits. Then, the results were analyzed micro-tomographically, histologically, and histomorphometrically. Our findings indicate that both scaffold architectures are biocompatible and support bone formation. The helical scaffolds, characterized by larger pore sizes and higher porosity, demonstrated significantly greater bone regeneration than the alternate structures. However, their lower mechanical strength presented limitations for use in load-bearing sites.
Collapse
Affiliation(s)
- Iván Alonso-Fernández
- Anatomy, Animal Production and Veterinary Clinical Sciences Department, Veterinary Faculty, Universidade de Santiago de Compostela, Campus Universitario s/n, 27002 Lugo, Spain; (M.L.-P.); (A.G.-C.); (F.M.-G.)
| | - Håvard Jostein Haugen
- Department of Biomaterials, Institute of Clinical Dentistry, Faculty of Dentistry, University of Oslo, 0317 Oslo, Norway; (H.J.H.); (L.P.N.)
| | - Liebert Parreiras Nogueira
- Department of Biomaterials, Institute of Clinical Dentistry, Faculty of Dentistry, University of Oslo, 0317 Oslo, Norway; (H.J.H.); (L.P.N.)
| | - Miriam López-Álvarez
- Centro de Investigación en Tecnologías, Energía y Procesos Industriales (CINTECX), Universidade de Vigo, Grupo de Novos Materiais, 36310 Vigo, Spain; (M.L.-Á.); (P.G.)
- Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO, 36213 Vigo, Spain
| | - Pío González
- Centro de Investigación en Tecnologías, Energía y Procesos Industriales (CINTECX), Universidade de Vigo, Grupo de Novos Materiais, 36310 Vigo, Spain; (M.L.-Á.); (P.G.)
- Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO, 36213 Vigo, Spain
| | - Mónica López-Peña
- Anatomy, Animal Production and Veterinary Clinical Sciences Department, Veterinary Faculty, Universidade de Santiago de Compostela, Campus Universitario s/n, 27002 Lugo, Spain; (M.L.-P.); (A.G.-C.); (F.M.-G.)
| | - Antonio González-Cantalapiedra
- Anatomy, Animal Production and Veterinary Clinical Sciences Department, Veterinary Faculty, Universidade de Santiago de Compostela, Campus Universitario s/n, 27002 Lugo, Spain; (M.L.-P.); (A.G.-C.); (F.M.-G.)
| | - Fernando Muñoz-Guzón
- Anatomy, Animal Production and Veterinary Clinical Sciences Department, Veterinary Faculty, Universidade de Santiago de Compostela, Campus Universitario s/n, 27002 Lugo, Spain; (M.L.-P.); (A.G.-C.); (F.M.-G.)
| |
Collapse
|
5
|
Li P, Dai J, Li Y, Alexander D, Čapek J, Geis-Gerstorfer J, Wan G, Han J, Yu Z, Li A. Zinc based biodegradable metals for bone repair and regeneration: Bioactivity and molecular mechanisms. Mater Today Bio 2024; 25:100932. [PMID: 38298560 PMCID: PMC10826336 DOI: 10.1016/j.mtbio.2023.100932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 12/12/2023] [Accepted: 12/25/2023] [Indexed: 02/02/2024] Open
Abstract
Bone fractures and critical-size bone defects are significant public health issues, and clinical treatment outcomes are closely related to the intrinsic properties of the utilized implant materials. Zinc (Zn)-based biodegradable metals (BMs) have emerged as promising bioactive materials because of their exceptional biocompatibility, appropriate mechanical properties, and controllable biodegradation. This review summarizes the state of the art in terms of Zn-based metals for bone repair and regeneration, focusing on bridging the gap between biological mechanism and required bioactivity. The molecular mechanism underlying the release of Zn ions from Zn-based BMs in the improvement of bone repair and regeneration is elucidated. By integrating clinical considerations and the specific bioactivity required for implant materials, this review summarizes the current research status of Zn-based internal fixation materials for promoting fracture healing, Zn-based scaffolds for regenerating critical-size bone defects, and Zn-based barrier membranes for reconstituting alveolar bone defects. Considering the significant progress made in the research on Zn-based BMs for potential clinical applications, the challenges and promising research directions are proposed and discussed.
Collapse
Affiliation(s)
- Ping Li
- Center of Oral Implantology, Stomatological Hospital, School of Stomatology, Southern Medical University, South Jiangnan Road No. 366, Guangzhou 510280, China
- School and Hospital of Stomatology, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Medical University, Guangzhou, Guangdong, 510182, China
- Department of Prosthodontics, School and Hospital of Stomatology, Guangzhou Medical University, Guangzhou, Guangdong, 510182, China
| | - Jingtao Dai
- Department of Orthodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, South Jiangnan Road No. 366, Guangzhou 510280, China
| | - Yageng Li
- Beijing Advanced Innovation Center for Materials Genome Engineering, School of Materials Science and Engineering, University of Science and Technology Beijing, Beijing 100083, China
| | - Dorothea Alexander
- Department of Oral and Maxillofacial Surgery, University Hospital Tübingen, Osianderstrasse 2-8, Tübingen 72076, Germany
| | - Jaroslav Čapek
- FZU – the Institute of Physics, Czech Academy of Sciences, Na Slovance 1999/2, Prague 8, 18200, Czech Republic
| | - Jürgen Geis-Gerstorfer
- Section Medical Materials Science and Technology, University Hospital Tübingen, Osianderstrasse 2-8, Tübingen 72076, Germany
| | - Guojiang Wan
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Jianmin Han
- National Engineering Laboratory for Digital and Material Technology of Stomatology, Department of Dental Materials, Peking University School and Hospital of Stomatology, Beijing 100081, China
| | - Zhentao Yu
- Institute of Advanced Wear & Corrosion Resistant and Functional Materials, Jinan University, Guangzhou 510632, China
| | - An Li
- Department of Periodontology, Stomatological Hospital, School of Stomatology, Southern Medical University, South Jiangnan Road 366, Guangzhou 510280, China
| |
Collapse
|
6
|
Chen S, Liu H, Wang Y, Wang S, Yang B, Sun D, Sun P. Overexpression of lncRNA LINC00665 inhibits the proliferation and chondroblast differentiation of bone marrow mesenchymal stem cells by targeting miR-214-3p. J Orthop Surg Res 2024; 19:2. [PMID: 38167456 PMCID: PMC10762961 DOI: 10.1186/s13018-023-04475-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 12/14/2023] [Indexed: 01/05/2024] Open
Abstract
BACKGROUND Osteoarthritis is a chronic disease mainly involving the damage of articular cartilage and the whole articular tissue, which is the main cause of disability in the elderly. To explore more effective treatment measures, this study analyzed the regulatory role and molecular mechanism of lncRNA LINC00665 (LINC00665) in the chondrogenic differentiation of bone marrow mesenchymal stem cells (BMSCs), providing a valuable theoretical basis for the pathogenesis and patient treatment of osteoarthritis. METHODS Osteoarthritis tissues and healthy tissues were obtained from 52 patients with osteoarthritis and 34 amputated patients without osteoarthritis, and the levels of LINC00665 and miR-214-3p were assessed by RT-qPCR. BMSCs were cultured and induced chondrogenic differentiation. The proliferation ability of BMSCs was detected by CCK-8 method, and the apoptosis level of BMSCs was evaluated by flow cytometry. The content of proteoglycan-glycosaminoglycan (GAG) in cartilage matrix was determined by Alcian blue staining. In addition, the binding relationship between LINC00665 and miR-214-3p was verified by luciferase reporter assay, and the molecular mechanism was further analyzed. RESULTS In osteoarthritis tissues, LINC00665 was elevated and miR-214-3p was down-regulated. With the chondrogenic differentiation of BMSCs, the level of GAG increased, and LINC00665 expression gradually decreased, while miR-214-3p level was on the contrary. After transfection of pcDNA3.1-LINC00665 in BMSCs, cell proliferation capacity was decreased, apoptosis rate was increased, and GAG content was reduced. Moreover, LINC00665 sponged miR-214-3p and negatively regulate its expression. Transfection of pcDNA3.1-LINC00665-miR-214-3p mimic changed the regulation of pcDNA3.1-LINC00665 on the viability and chondrogenic differentiation of BMSCs. CONCLUSIONS Overexpression of lncRNA LINC00665 inhibited the proliferation and chondrogenic differentiation of BMSCs by targeting miR-214-3p. The LINC00665/miR-214-3p axis may improve joint damage and alleviate the progression of osteoarthritis.
Collapse
Affiliation(s)
- Siyuan Chen
- Surgery of Spinal Degeneration and Deformity, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524000, China
| | - Hui Liu
- Department of Nursing, Peking University Third Hospital Qinhuangdao Hospital, Qinhuangdao, 066000, China
| | - Yue Wang
- Department of Nursing, Peking University Third Hospital Qinhuangdao Hospital, Qinhuangdao, 066000, China
| | - Shuyuan Wang
- Department of Nursing, Peking University Third Hospital Qinhuangdao Hospital, Qinhuangdao, 066000, China
| | - Bo Yang
- Department of Nursing, Peking University Third Hospital Qinhuangdao Hospital, Qinhuangdao, 066000, China
| | - Di Sun
- Department of Orthopedics, Peking University Third Hospital Qinhuangdao Hospital, Qinhuangdao, 066000, China
| | - Pengxiao Sun
- First Department of Joint, Xi'an International Medical Center Hospital, No.777, Xitai Road, Gaoxin District, Xi'an, 710000, China.
| |
Collapse
|
7
|
Garot C, Schoffit S, Monfoulet C, Machillot P, Deroy C, Roques S, Vial J, Vollaire J, Renard M, Ghanem H, El‐Hafci H, Decambron A, Josserand V, Bordenave L, Bettega G, Durand M, Manassero M, Viateau V, Logeart‐Avramoglou D, Picart C. 3D-Printed Osteoinductive Polymeric Scaffolds with Optimized Architecture to Repair a Sheep Metatarsal Critical-Size Bone Defect. Adv Healthc Mater 2023; 12:e2301692. [PMID: 37655491 PMCID: PMC11468956 DOI: 10.1002/adhm.202301692] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 08/10/2023] [Indexed: 09/02/2023]
Abstract
The reconstruction of critical-size bone defects in long bones remains a challenge for clinicians. A new osteoinductive medical device is developed here for long bone repair by combining a 3D-printed architectured cylindrical scaffold made of clinical-grade polylactic acid (PLA) with a polyelectrolyte film coating delivering the osteogenic bone morphogenetic protein 2 (BMP-2). This film-coated scaffold is used to repair a sheep metatarsal 25-mm long critical-size bone defect. In vitro and in vivo biocompatibility of the film-coated PLA material is proved according to ISO standards. Scaffold geometry is found to influence BMP-2 incorporation. Bone regeneration is followed using X-ray scans, µCT scans, and histology. It is shown that scaffold internal geometry, notably pore shape, influenced bone regeneration, which is homogenous longitudinally. Scaffolds with cubic pores of ≈870 µm and a low BMP-2 dose of ≈120 µg cm-3 induce the best bone regeneration without any adverse effects. The visual score given by clinicians during animal follow-up is found to be an easy way to predict bone regeneration. This work opens perspectives for a clinical application in personalized bone regeneration.
Collapse
Affiliation(s)
- Charlotte Garot
- CNRS EMR 5000 Biomimetism and Regenerative Medicine (BRM)INSERM U1292 BiosantéCEAUniversité Grenoble Alpes17 avenue des MartyrsGrenobleF‐38054France
| | - Sarah Schoffit
- Ecole Nationale Vétérinaire d'AlfortUniversité Paris‐EstMaisons‐AlfortF‐94704France
- CNRSINSERMENVAB3OAUniversité Paris CitéParisF‐75010France
| | - Cécile Monfoulet
- INSERMInstitut BergoniéUniversity of BordeauxCIC 1401BordeauxF‐33000France
- CIC‐ITINSERMInstitut BergoniéCHU de BordeauxCIC 1401BordeauxF‐33000France
| | - Paul Machillot
- CNRS EMR 5000 Biomimetism and Regenerative Medicine (BRM)INSERM U1292 BiosantéCEAUniversité Grenoble Alpes17 avenue des MartyrsGrenobleF‐38054France
| | - Claire Deroy
- INSERMInstitut BergoniéUniversity of BordeauxCIC 1401BordeauxF‐33000France
- CIC‐ITINSERMInstitut BergoniéCHU de BordeauxCIC 1401BordeauxF‐33000France
| | - Samantha Roques
- INSERMInstitut BergoniéUniversity of BordeauxCIC 1401BordeauxF‐33000France
- CIC‐ITINSERMInstitut BergoniéCHU de BordeauxCIC 1401BordeauxF‐33000France
| | - Julie Vial
- Ecole Nationale Vétérinaire d'AlfortUniversité Paris‐EstMaisons‐AlfortF‐94704France
- CNRSINSERMENVAB3OAUniversité Paris CitéParisF‐75010France
| | - Julien Vollaire
- INSERM U1209Institute of Advanced BiosciencesGrenobleF‐38000France
- Institute of Advanced BiosciencesUniversité Grenoble AlpesGrenobleF‐38000France
| | - Martine Renard
- INSERMInstitut BergoniéUniversity of BordeauxCIC 1401BordeauxF‐33000France
- CIC‐ITINSERMInstitut BergoniéCHU de BordeauxCIC 1401BordeauxF‐33000France
| | - Hasan Ghanem
- CNRSINSERMENVAB3OAUniversité Paris CitéParisF‐75010France
| | | | - Adeline Decambron
- Ecole Nationale Vétérinaire d'AlfortUniversité Paris‐EstMaisons‐AlfortF‐94704France
- CNRSINSERMENVAB3OAUniversité Paris CitéParisF‐75010France
| | - Véronique Josserand
- INSERM U1209Institute of Advanced BiosciencesGrenobleF‐38000France
- Institute of Advanced BiosciencesUniversité Grenoble AlpesGrenobleF‐38000France
| | - Laurence Bordenave
- INSERMInstitut BergoniéUniversity of BordeauxCIC 1401BordeauxF‐33000France
- CIC‐ITINSERMInstitut BergoniéCHU de BordeauxCIC 1401BordeauxF‐33000France
| | - Georges Bettega
- INSERM U1209Institute of Advanced BiosciencesGrenobleF‐38000France
- Service de Chirurgie Maxillo‐FacialeCentre Hospitalier Annecy Genevois1 avenue de l'hôpitalEpagny Metz‐TessyF‐74370France
| | - Marlène Durand
- INSERMInstitut BergoniéUniversity of BordeauxCIC 1401BordeauxF‐33000France
- CIC‐ITINSERMInstitut BergoniéCHU de BordeauxCIC 1401BordeauxF‐33000France
| | - Mathieu Manassero
- Ecole Nationale Vétérinaire d'AlfortUniversité Paris‐EstMaisons‐AlfortF‐94704France
- CNRSINSERMENVAB3OAUniversité Paris CitéParisF‐75010France
| | - Véronique Viateau
- Ecole Nationale Vétérinaire d'AlfortUniversité Paris‐EstMaisons‐AlfortF‐94704France
- CNRSINSERMENVAB3OAUniversité Paris CitéParisF‐75010France
| | | | - Catherine Picart
- CNRS EMR 5000 Biomimetism and Regenerative Medicine (BRM)INSERM U1292 BiosantéCEAUniversité Grenoble Alpes17 avenue des MartyrsGrenobleF‐38054France
- Institut Universitaire de France (IUF)1 rue DescartesParis CEDEX 0575231France
| |
Collapse
|
8
|
Liang QL, Xu HG, Yu L, Ding MR, Li YT, Qi GF, Zhang K, Wang L, Wang H, Cui X. Binding-induced fibrillogenesis peptide inhibits RANKL-mediated osteoclast activation against osteoporosis. Biomaterials 2023; 302:122331. [PMID: 37741149 DOI: 10.1016/j.biomaterials.2023.122331] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 09/05/2023] [Accepted: 09/12/2023] [Indexed: 09/25/2023]
Abstract
Osteoporosis is primarily driven by an imbalance between bone resorption and formation, stemming from enhanced osteoclast activity during bone remodeling. At the crux of this mechanism lies the pivotal RANK-RANKL-OPG axis. In our study, we designed two binding-induced fibrillogenesis (BIF) peptides, namely BIFP and BIFY, targeting RANK and RANKL, respectively. These BIF peptides, with distinct hydrophilic and hydrophobic characteristics, assemble into nanoparticles (NPs) in aqueous solution. Through specific ligand-receptor interactions, these NPs efficiently target and bind to specific proteins, resulting in the formation of fibrous networks that effectively inhibit the RANK-RANKL associations. Experiments have confirmed the potent inhibitory effects of peptides on both osteoclast differentiation and function. Compared with the +RANKL controls, BIFP and BIFY demonstrated a more remarkable reduction in tartrate resistant acid phosphatase (TRAP)-positive cells, achieving an impressive decline of 82.8% and 70.7%, respectively. Remarkably, the administration of BIFP led to a substantial reduction in bone resorption pit area by 17.4%, compared to a significant increase of 92.4% in the +RANKL groups. In vivo experiments on an ovariectomized mouse model demonstrated that the BIFP treated group exhibited an impressive 2.6-fold elevation in bone mineral density and an astounding 4.0-fold enhancement in bone volume/total volume as against those of the PBS-treated group. Overall, BIF peptides demonstrate remarkable abilities to impede osteoclast differentiation, presenting promising prospects for the treatment of osteoporosis.
Collapse
Affiliation(s)
- Qi-Lin Liang
- College of Medicine, Southwest Jiaotong University, No. 111 Beiyiduan, Second Ring Road, Chengdu, 610031, Sichuan Province, China; CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, Beijing, 100190, China
| | - Huan-Ge Xu
- College of Medicine, Southwest Jiaotong University, No. 111 Beiyiduan, Second Ring Road, Chengdu, 610031, Sichuan Province, China; CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, Beijing, 100190, China
| | - Long Yu
- Department of Orthopaedics, The 4th Medical Center of Chinese PLA General Hospital, Jia No.17 Heishanhu Road, Beijing, 100091, China
| | - Meng-Ru Ding
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, Beijing, 100190, China
| | - Yu-Ting Li
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, Beijing, 100190, China
| | - Gao-Feng Qi
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, Beijing, 100190, China
| | - Kuo Zhang
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, Beijing, 100190, China
| | - Lei Wang
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, Beijing, 100190, China.
| | - Hao Wang
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, Beijing, 100190, China.
| | - Xu Cui
- Department of Orthopaedics, The 4th Medical Center of Chinese PLA General Hospital, Jia No.17 Heishanhu Road, Beijing, 100091, China.
| |
Collapse
|
9
|
Alonso-Fernández I, Haugen HJ, López-Peña M, González-Cantalapiedra A, Muñoz F. Use of 3D-printed polylactic acid/bioceramic composite scaffolds for bone tissue engineering in preclinical in vivo studies: A systematic review. Acta Biomater 2023; 168:1-21. [PMID: 37454707 DOI: 10.1016/j.actbio.2023.07.013] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 07/11/2023] [Accepted: 07/12/2023] [Indexed: 07/18/2023]
Abstract
3D-printed composite scaffolds have emerged as an alternative to deal with existing limitations when facing bone reconstruction. The aim of the study was to systematically review the feasibility of using PLA/bioceramic composite scaffolds manufactured by 3D-printing technologies as bone grafting materials in preclinical in vivo studies. Electronic databases were searched using specific search terms, and thirteen manuscripts were selected after screening. The synthesis of the scaffolds was carried out using mainly extrusion-based techniques. Likewise, hydroxyapatite was the most used bioceramic for synthesizing composites with a PLA matrix. Among the selected studies, seven were conducted in rats and six in rabbits, but the high variability that exists regarding the experimental process made it difficult to compare them. Regarding the results, PLA/Bioceramic composite scaffolds have shown to be biocompatible and mechanically resistant. Preclinical studies elucidated the ability of the scaffolds to be used as bone grafts, allowing bone growing without adverse reactions. In conclusion, PLA/Bioceramics scaffolds have been demonstrated to be a promising alternative for treating bone defects. Nevertheless, more care should be taken when designing and performing in vivo trials, since the lack of standardization of the processes, which prevents the comparison of the results and reduces the quality of the information. STATEMENT OF SIGNIFICANCE: 3D-printed polylactic acid/bioceramic composite scaffolds have emerged as an alternative to deal with existing limitations when facing bone reconstruction. Since preclinical in vivo studies with animal models represent a mandatory step for clinical translation, the present manuscript analyzed and discussed not only those aspects related to the selection of the bioceramic material, the synthesis of the implants and their characterization. But provides a new approach to understand how the design and perform of clinical trials, as well as the selection of the analysis methods, may affect the obtained results, by covering authors' knowledgebase from veterinary medicine to biomaterial science. Thus, this study aims to systematically review the feasibility of using polylactic acid/bioceramic scaffolds as grafting materials in preclinical trials.
Collapse
Affiliation(s)
- Iván Alonso-Fernández
- Anatomy, Animal Production and Veterinary Clinical Sciences Department, Veterinary Faculty, Universidade de Santiago de Compostela, Campus Universitario s/n, 27002 Lugo, Spain.
| | - Håvard Jostein Haugen
- Department of Biomaterials, Institute of Clinical Dentistry, Faculty of Dentistry, University of Oslo, Oslo, Norway
| | - Mónica López-Peña
- Anatomy, Animal Production and Veterinary Clinical Sciences Department, Veterinary Faculty, Universidade de Santiago de Compostela, Campus Universitario s/n, 27002 Lugo, Spain
| | - Antonio González-Cantalapiedra
- Anatomy, Animal Production and Veterinary Clinical Sciences Department, Veterinary Faculty, Universidade de Santiago de Compostela, Campus Universitario s/n, 27002 Lugo, Spain
| | - Fernando Muñoz
- Anatomy, Animal Production and Veterinary Clinical Sciences Department, Veterinary Faculty, Universidade de Santiago de Compostela, Campus Universitario s/n, 27002 Lugo, Spain
| |
Collapse
|
10
|
Guizzardi S, Colangelo MT, Mirandola P, Galli C. Modeling new trends in bone regeneration, using the BERTopic approach. Regen Med 2023; 18:719-734. [PMID: 37577987 DOI: 10.2217/rme-2023-0096] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/15/2023] Open
Abstract
Aim: Bibliometric surveys are time-consuming endeavors, which cannot be scaled up to meet the challenges of ever-expanding fields, such as bone regeneration. Artificial intelligence, however, can provide smart tools to screen massive amounts of literature, and we relied on this technology to automatically identify research topics. Materials & methods: We used the BERTopic algorithm to detect the topics in a corpus of MEDLINE manuscripts, mapping their similarities and highlighting research hotspots. Results: Using BERTopic, we identified 372 topics and were able to assess the growing importance of innovative and recent fields of investigation such as 3D printing and extracellular vescicles. Conclusion: BERTopic appears as a suitable tool to set up automatic screening routines to track the progress in bone regeneration.
Collapse
Affiliation(s)
- Stefano Guizzardi
- Department of Medicine & Surgery, Histology & Embryology Lab, University of Parma, Parma, 43126, Italy
| | - Maria Teresa Colangelo
- Department of Medicine & Surgery, Histology & Embryology Lab, University of Parma, Parma, 43126, Italy
| | - Prisco Mirandola
- Department of Medicine & Surgery, Histology & Embryology Lab, University of Parma, Parma, 43126, Italy
| | - Carlo Galli
- Department of Medicine & Surgery, Histology & Embryology Lab, University of Parma, Parma, 43126, Italy
| |
Collapse
|
11
|
Feng B, Zhang M, Qin C, Zhai D, Wang Y, Zhou Y, Chang J, Zhu Y, Wu C. 3D printing of conch-like scaffolds for guiding cell migration and directional bone growth. Bioact Mater 2023; 22:127-140. [PMID: 36203957 PMCID: PMC9525999 DOI: 10.1016/j.bioactmat.2022.09.014] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Revised: 09/09/2022] [Accepted: 09/15/2022] [Indexed: 11/05/2022] Open
Abstract
Regeneration of severe bone defects remains an enormous challenge in clinic. Developing regenerative scaffolds to directionally guide bone growth is a potential strategy to overcome this hurdle. Conch, an interesting creature widely spreading in ocean, has tough spiral shell that can continuously grow along the spiral direction. Herein, inspired by the physiological features of conches, a conch-like (CL) scaffold based on β-TCP bioceramic material was successfully prepared for guiding directional bone growth via digital light processing (DLP)-based 3D printing. Benefiting from the spiral structure, the CL scaffolds significantly improved cell adhesion, proliferation and osteogenic differentiation in vitro compared to the conventional 3D scaffolds. Particularly, the spiral structure in the scaffolds could efficiently induce cells to migrate from the bottom to the top of the scaffolds, which was like “cells climbing stairs”. Furthermore, the capability of guiding directional bone growth for the CL scaffolds was demonstrated by a special half-embedded femoral defects model in rabbits. The new bone tissue could consecutively grow into the protruded part of the scaffolds along the spiral cavities. This work provides a promising strategy to construct biomimetic biomaterials for guiding directional bone tissue growth, which offers a new treatment concept for severe bone defects, and even limb regeneration. A conch-like scaffold was firstly developed for guiding directional bone growth. The CL scaffolds efficiently induced cells “climbing stairs”- like-migrating. The CL scaffolds showed improved bioactivities benefited from the spiral structure. This work provided a new treatment concept for severe bone defects.
Collapse
|
12
|
Chauhan A, Bhatt AD. A review on design of scaffold for osteoinduction: Toward the unification of independent design variables. Biomech Model Mechanobiol 2023; 22:1-21. [PMID: 36121530 DOI: 10.1007/s10237-022-01635-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 09/05/2022] [Indexed: 11/29/2022]
Abstract
Biophysical stimulus quantifies the osteoinductivity of the scaffold concerning the mechanoregulatory mathematical models of scaffold-assisted cellular differentiation. Consider a set of independent structural variables ($) that comprises bulk porosity levels ([Formula: see text]) and a set of morphological features of the micro-structure ([Formula: see text]) associated with scaffolds, i.e., [Formula: see text]. The literature suggests that biophysical stimulus ([Formula: see text]) is a function of independent structural variables ($). Limited understanding of the functional correlation between biophysical stimulus and structural features results in the lack of the desired osteoinductivity in a scaffold. Consequently, it limits their broad applicability to assist bone tissue regeneration for treating critical-sized bone fractures. The literature indicates the existence of multi-dimensional independent design variable space as a probable reason for the general lack of osteoinductivity in scaffolds. For instance, known morphological features are the size, shape, orientation, continuity, and connectivity of the porous regions in the scaffold. It implies that the number of independent variables ([Formula: see text]) is more than two, i.e., [Formula: see text], which interact and influence the magnitude of [Formula: see text] in a unified manner. The efficiency of standard engineering design procedures to analyze the correlation between dependent variable ([Formula: see text]) and independent variables ($) in 3D mutually orthogonal Cartesian coordinate system diminishes proportionally with the increase in the number of independent variables ([Formula: see text]) (Deb in Optimization for engineering design-algorithms and examples, PHI Learning Private Limited, New Delhi, 2012). Therefore, there is an immediate need to devise a framework that has the potential to quantify the micro-structural's morphological features in a unified manner to increase the prospects of scaffold-assisted bone tissue regeneration.
Collapse
Affiliation(s)
- Atul Chauhan
- Department of Mechanical Engineering, Motilal Nehru National Institute of Technology Allahabad, Prayagraj, Uttar Pradesh, 211004, India.
| | - Amba D Bhatt
- Department of Mechanical Engineering, Motilal Nehru National Institute of Technology Allahabad, Prayagraj, Uttar Pradesh, 211004, India
| |
Collapse
|
13
|
Nedelcu L, Ferreira JMF, Popa AC, Amarande L, Nan B, Bălescu LM, Geambașu CD, Cioangher MC, Leonat L, Grigoroscuță M, Cristea D, Stroescu H, Ciocoiu RC, Stan GE. Multi-Parametric Exploration of a Selection of Piezoceramic Materials for Bone Graft Substitute Applications. MATERIALS (BASEL, SWITZERLAND) 2023; 16:901. [PMID: 36769908 PMCID: PMC9917895 DOI: 10.3390/ma16030901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 01/11/2023] [Accepted: 01/13/2023] [Indexed: 06/18/2023]
Abstract
This work was devoted to the first multi-parametric unitary comparative analysis of a selection of sintered piezoceramic materials synthesised by solid-state reactions, aiming to delineate the most promising biocompatible piezoelectric material, to be further implemented into macro-porous ceramic scaffolds fabricated by 3D printing technologies. The piezoceramics under scrutiny were: KNbO3, LiNbO3, LiTaO3, BaTiO3, Zr-doped BaTiO3, and the (Ba0.85Ca0.15)(Ti0.9Zr0.1)O3 solid solution (BCTZ). The XRD analysis revealed the high crystallinity of all sintered ceramics, while the best densification was achieved for the BaTiO3-based materials via conventional sintering. Conjunctively, BCTZ yielded the best combination of functional properties-piezoelectric response (in terms of longitudinal piezoelectric constant and planar electromechanical coupling factor) and mechanical and in vitro osteoblast cell compatibility. The selected piezoceramic was further used as a base material for the robocasting fabrication of 3D macro-porous scaffolds (porosity of ~50%), which yielded a promising compressive strength of ~20 MPa (higher than that of trabecular bone), excellent cell colonization capability, and noteworthy cytocompatibility in osteoblast cell cultures, analogous to the biological control. Thereby, good prospects for the possible development of a new generation of synthetic bone graft substitutes endowed with the piezoelectric effect as a stimulus for the enhancement of osteogenic capacity were settled.
Collapse
Affiliation(s)
- Liviu Nedelcu
- National Institute of Materials Physics, 077125 Magurele, Romania
| | - José M. F. Ferreira
- Department of Materials and Ceramic Engineering, CICECO—Aveiro Materials Institute, University of Aveiro, 3810-193 Aveiro, Portugal
| | | | | | - Bo Nan
- Department of Materials and Ceramic Engineering, CICECO—Aveiro Materials Institute, University of Aveiro, 3810-193 Aveiro, Portugal
| | | | | | | | - Lucia Leonat
- National Institute of Materials Physics, 077125 Magurele, Romania
| | | | - Daniel Cristea
- Department of Materials Science, Faculty of Materials Science and Engineering, Transilvania University of Brasov, 500068 Brasov, Romania
| | - Hermine Stroescu
- “Ilie Murgulescu” Institute of Physical Chemistry of the Romanian Academy, 060021 Bucharest, Romania
| | - Robert Cătălin Ciocoiu
- Department of Metallic Materials Science, Physical Metallurgy, University Politehnica of Bucharest, 060042 Bucharest, Romania
| | - George E. Stan
- National Institute of Materials Physics, 077125 Magurele, Romania
| |
Collapse
|
14
|
Ng WC, Lokanathan Y, Baki MM, Fauzi MB, Zainuddin AA, Azman M. Tissue Engineering as a Promising Treatment for Glottic Insufficiency: A Review on Biomolecules and Cell-Laden Hydrogel. Biomedicines 2022; 10:3082. [PMID: 36551838 PMCID: PMC9775346 DOI: 10.3390/biomedicines10123082] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 11/11/2022] [Accepted: 11/16/2022] [Indexed: 12/03/2022] Open
Abstract
Glottic insufficiency is widespread in the elderly population and occurs as a result of secondary damage or systemic disease. Tissue engineering is a viable treatment for glottic insufficiency since it aims to restore damaged nerve tissue and revitalize aging muscle. After injection into the biological system, injectable biomaterial delivers cost- and time-effectiveness while acting as a protective shield for cells and biomolecules. This article focuses on injectable biomaterials that transport cells and biomolecules in regenerated tissue, particularly adipose, muscle, and nerve tissue. We propose Wharton's Jelly mesenchymal stem cells (WJMSCs), induced pluripotent stem cells (IP-SCs), basic fibroblast growth factor (bFGF), vascular endothelial growth factor (VEGF), hepatocyte growth factor (HGF), insulin growth factor-1 (IGF-1) and extracellular vesicle (EV) as potential cells and macromolecules to be included into biomaterials, with some particular testing to support them as a promising translational medicine for vocal fold regeneration.
Collapse
Affiliation(s)
- Wan-Chiew Ng
- Department of Otorhinolaryngology-Head and Neck Surgery, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
| | - Yogeswaran Lokanathan
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
| | - Marina Mat Baki
- Department of Otorhinolaryngology-Head and Neck Surgery, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
| | - Mh Busra Fauzi
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
| | - Ani Amelia Zainuddin
- Department of Obstetrics and Gynaecology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
| | - Mawaddah Azman
- Department of Otorhinolaryngology-Head and Neck Surgery, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
| |
Collapse
|
15
|
Paladini F, Pollini M. Novel Approaches and Biomaterials for Bone Tissue Engineering: A Focus on Silk Fibroin. MATERIALS (BASEL, SWITZERLAND) 2022; 15:6952. [PMID: 36234293 PMCID: PMC9572978 DOI: 10.3390/ma15196952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 09/27/2022] [Accepted: 10/04/2022] [Indexed: 05/16/2023]
Abstract
Bone tissue engineering (BTE) represents a multidisciplinary research field involving many aspects of biology, engineering, material science, clinical medicine and genetics to create biological substitutes to promote bone regeneration. The definition of the most appropriate biomaterials and structures for BTE is still a challenge for researchers, aiming at simultaneously combining different features such as tissue generation properties, biocompatibility, porosity and mechanical strength. In this scenario, among the biomaterials for BTE, silk fibroin represents a valuable option for the development of functional devices because of its unique biological properties and the multiple chances of processing. This review article aims at providing the reader with a general overview of the most recent progresses in bone tissue engineering in terms of approaches and materials with a special focus on silk fibroin and the related mechanisms involved in bone regeneration, and presenting interesting results obtained by different research groups, which assessed the great potential of this protein for bone tissue engineering.
Collapse
Affiliation(s)
- Federica Paladini
- Department of Engineering for Innovation, University of Salento, Via Monteroni, 73100 Lecce, Italy
- Caresilk S.r.l.s., Via Monteroni c/o Technological District DHITECH, 73100 Lecce, Italy
| | - Mauro Pollini
- Department of Engineering for Innovation, University of Salento, Via Monteroni, 73100 Lecce, Italy
- Caresilk S.r.l.s., Via Monteroni c/o Technological District DHITECH, 73100 Lecce, Italy
| |
Collapse
|
16
|
Tian B, Li X, Zhang J, Zhang M, Gan D, Deng D, Sun L, He X, Wu C, Chen F. A 3D-printed molybdenum-containing scaffold exerts dual pro-osteogenic and anti-osteoclastogenic effects to facilitate alveolar bone repair. Int J Oral Sci 2022; 14:45. [PMID: 36064833 PMCID: PMC9445063 DOI: 10.1038/s41368-022-00195-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 07/12/2022] [Accepted: 08/02/2022] [Indexed: 11/19/2022] Open
Abstract
The positive regulation of bone-forming osteoblast activity and the negative feedback regulation of osteoclastic activity are equally important in strategies to achieve successful alveolar bone regeneration. Here, a molybdenum (Mo)-containing bioactive glass ceramic scaffold with solid-strut-packed structures (Mo-scaffold) was printed, and its ability to regulate pro-osteogenic and anti-osteoclastogenic cellular responses was evaluated in vitro and in vivo. We found that extracts derived from Mo-scaffold (Mo-extracts) strongly stimulated osteogenic differentiation of bone marrow mesenchymal stem cells and inhibited differentiation of osteoclast progenitors. The identified comodulatory effect was further demonstrated to arise from Mo ions in the Mo-extract, wherein Mo ions suppressed osteoclastic differentiation by scavenging reactive oxygen species (ROS) and inhibiting mitochondrial biogenesis in osteoclasts. Consistent with the in vitro findings, the Mo-scaffold was found to significantly promote osteoblast-mediated bone formation and inhibit osteoclast-mediated bone resorption throughout the bone healing process, leading to enhanced bone regeneration. In combination with our previous finding that Mo ions participate in material-mediated immunomodulation, this study offers the new insight that Mo ions facilitate bone repair by comodulating the balance between bone formation and resorption. Our findings suggest that Mo ions are multifunctional cellular modulators that can potentially be used in biomaterial design and bone tissue engineering.
Collapse
Affiliation(s)
- Beimin Tian
- Department of Periodontology, State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases, Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, School of Stomatology, Fourth Military Medical University, Xi'an, China
| | - Xuan Li
- Department of Periodontology, State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases, Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, School of Stomatology, Fourth Military Medical University, Xi'an, China
| | - Jiujiu Zhang
- Department of Periodontology, State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases, Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, School of Stomatology, Fourth Military Medical University, Xi'an, China
| | - Meng Zhang
- State Key Laboratory of High-Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, China
| | - Dian Gan
- Department of Periodontology, State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases, Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, School of Stomatology, Fourth Military Medical University, Xi'an, China
| | - Daokun Deng
- Department of Periodontology, State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases, Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, School of Stomatology, Fourth Military Medical University, Xi'an, China
| | - Lijuan Sun
- Department of Periodontology, State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases, Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, School of Stomatology, Fourth Military Medical University, Xi'an, China
| | - Xiaotao He
- Department of Periodontology, State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases, Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, School of Stomatology, Fourth Military Medical University, Xi'an, China
| | - Chengtie Wu
- State Key Laboratory of High-Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, China.
| | - Faming Chen
- Department of Periodontology, State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases, Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, School of Stomatology, Fourth Military Medical University, Xi'an, China.
| |
Collapse
|
17
|
Gabetti S, Masante B, Cochis A, Putame G, Sanginario A, Armando I, Fiume E, Scalia AC, Daou F, Baino F, Salati S, Morbiducci U, Rimondini L, Bignardi C, Massai D. An automated 3D-printed perfusion bioreactor combinable with pulsed electromagnetic field stimulators for bone tissue investigations. Sci Rep 2022; 12:13859. [PMID: 35974079 PMCID: PMC9381575 DOI: 10.1038/s41598-022-18075-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 08/04/2022] [Indexed: 11/19/2022] Open
Abstract
In bone tissue engineering research, bioreactors designed for replicating the main features of the complex native environment represent powerful investigation tools. Moreover, when equipped with automation, their use allows reducing user intervention and dependence, increasing reproducibility and the overall quality of the culture process. In this study, an automated uni-/bi-directional perfusion bioreactor combinable with pulsed electromagnetic field (PEMF) stimulation for culturing 3D bone tissue models is proposed. A user-friendly control unit automates the perfusion, minimizing the user dependency. Computational fluid dynamics simulations supported the culture chamber design and allowed the estimation of the shear stress values within the construct. Electromagnetic field simulations demonstrated that, in case of combination with a PEMF stimulator, the construct can be exposed to uniform magnetic fields. Preliminary biological tests on 3D bone tissue models showed that perfusion promotes the release of the early differentiation marker alkaline phosphatase. The histological analysis confirmed that perfusion favors cells to deposit more extracellular matrix (ECM) with respect to the static culture and revealed that bi-directional perfusion better promotes ECM deposition across the construct with respect to uni-directional perfusion. Lastly, the Real-time PCR results of 3D bone tissue models cultured under bi-directional perfusion without and with PEMF stimulation revealed that the only perfusion induced a ~ 40-fold up-regulation of the expression of the osteogenic gene collagen type I with respect to the static control, while a ~ 80-fold up-regulation was measured when perfusion was combined with PEMF stimulation, indicating a positive synergic pro-osteogenic effect of combined physical stimulations.
Collapse
Affiliation(s)
- Stefano Gabetti
- PolitoBIOMed Lab, Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy
- Interuniversity Center for the Promotion of the 3Rs Principles in Teaching and Research, Turin, Italy
| | - Beatrice Masante
- PolitoBIOMed Lab, Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy
| | - Andrea Cochis
- Laboratory of Biomedical Materials, Center for Translational Research on Autoimmune and Allergic Disease-CAAD, Department of Health Sciences, University of Piemonte Orientale UPO, Novara, Italy
| | - Giovanni Putame
- PolitoBIOMed Lab, Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy
- Interuniversity Center for the Promotion of the 3Rs Principles in Teaching and Research, Turin, Italy
| | - Alessandro Sanginario
- Department of Electronics and Telecommunications, Politecnico di Torino, Turin, Italy
| | - Ileana Armando
- PolitoBIOMed Lab, Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy
- Department of Information Engineering, University of Brescia, Brescia, Italy
| | - Elisa Fiume
- Department of Applied Science and Technology, Politecnico di Torino, Turin, Italy
| | - Alessandro Calogero Scalia
- Laboratory of Biomedical Materials, Center for Translational Research on Autoimmune and Allergic Disease-CAAD, Department of Health Sciences, University of Piemonte Orientale UPO, Novara, Italy
| | - Farah Daou
- Laboratory of Biomedical Materials, Center for Translational Research on Autoimmune and Allergic Disease-CAAD, Department of Health Sciences, University of Piemonte Orientale UPO, Novara, Italy
| | - Francesco Baino
- Department of Applied Science and Technology, Politecnico di Torino, Turin, Italy
| | | | - Umberto Morbiducci
- PolitoBIOMed Lab, Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy
- Interuniversity Center for the Promotion of the 3Rs Principles in Teaching and Research, Turin, Italy
| | - Lia Rimondini
- Laboratory of Biomedical Materials, Center for Translational Research on Autoimmune and Allergic Disease-CAAD, Department of Health Sciences, University of Piemonte Orientale UPO, Novara, Italy
| | - Cristina Bignardi
- PolitoBIOMed Lab, Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy
- Interuniversity Center for the Promotion of the 3Rs Principles in Teaching and Research, Turin, Italy
| | - Diana Massai
- PolitoBIOMed Lab, Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy.
- Interuniversity Center for the Promotion of the 3Rs Principles in Teaching and Research, Turin, Italy.
| |
Collapse
|
18
|
Notoginsenoside R1 Promotes Migration, Adhesin, Spreading, and Osteogenic Differentiation of Human Adipose Tissue-Derived Mesenchymal Stromal Cells. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27113403. [PMID: 35684342 PMCID: PMC9182421 DOI: 10.3390/molecules27113403] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 05/20/2022] [Accepted: 05/21/2022] [Indexed: 11/18/2022]
Abstract
Cellular activities, such as attachment, spreading, proliferation, migration, and differentiation are indispensable for the success of bone tissue engineering. Mesenchymal stromal cells (MSCs) are the key precursor cells to regenerate bone. Bioactive compounds from natural products had shown bone regenerative potential. Notoginsenoside R1 (NGR1) is a primary bioactive natural compound that regulates various biological activities, including cardiovascular protection, neuro-protection, and anti-cancer effects. However, the effect of NGR1 on migration, adhesion, spreading, and osteogenic differentiation of MSCs required for bone tissue engineering application has not been tested properly. In this study, we aimed to analyze the effect of NGR1 on the cellular activities of MSCs. Since human adipose-derived stromal cells (hASCs) are commonly used MSCs for bone tissue engineering, we used hASCs as a model of MSCs. The optimal concentration of 0.05 μg/mL NGR1 was biocompatible and promoted migration and osteogenic differentiation of hASCs. Pro-angiogenic factor VEGF expression was upregulated in NGR1-treated hASCs. NGR1 enhanced the adhesion and spreading of hASCs on the bio-inert glass surface. NGR1 robustly promoted hASCs adhesion and survival in 3D-printed TCP scaffold both in vitro and in vivo. NGR1 mitigated LPS-induced expression of inflammatory markers IL-1β, IL-6, and TNF-α in hASCs as well as inhibited the RANKL/OPG expression ratio. In conclusion, the biocompatible NGR1 promoted the migration, adhesion, spreading, osteogenic differentiation, and anti-inflammatory properties of hASCs.
Collapse
|