1
|
Wang M, Xu Y, Cao L, Xiong L, Shang D, Cong Y, Zhao D, Wei X, Li J, Fu D, Lian H, Zhao Z. Mechanical and biological properties of 3D printed bone tissue engineering scaffolds. Front Bioeng Biotechnol 2025; 13:1545693. [PMID: 40260017 PMCID: PMC12010109 DOI: 10.3389/fbioe.2025.1545693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Accepted: 03/10/2025] [Indexed: 04/23/2025] Open
Abstract
Bone defects have historically represented a significant challenge in clinical practice, with traditional surgical intervention remaining the gold standard for their management. However, due to the problem of the origin of autologous and allogeneic bone and the complex and diverse bone defects, traditional surgical methods sometimes cannot meet the treatment needs and expectations of patients. The development of bone tissue engineering and 3D printing technology provides new ideas for bone defect repair. Ideal bioscaffold materials must have good mechanical properties, biocompatibility, osteoinduction and bone conduction capabilities. Additionally, factors such as degradation rate, appropriate porosity and a sustained antibacterial effect must be taken into account. The combination of 3D printing technology and synthetic composite biomaterial scaffolds has become a well-established approach in the treatment of complex bone defects, offering innovative solutions for bone defect repair. The combined application of seed cells, signalling factors and biological scaffolds is also beneficial to improve the therapeutic effect of complex bone defects. This article will therefore examine some of the most commonly used 3D printing technologies for biological scaffolds and the most prevalent bioscaffold materials suitable for 3D printing. An analysis will be conducted on the mechanical and biological properties of these materials to elucidate their respective advantages and limitations.
Collapse
Affiliation(s)
- Mingxuan Wang
- Orthopaedic Department, Affiliated ZhongShan Hospital of Dalian University, Dalian, Liaoning, China
| | - Yunpeng Xu
- Orthopaedic Department, Affiliated ZhongShan Hospital of Dalian University, Dalian, Liaoning, China
| | - Luoxi Cao
- Orthopaedic Department, Affiliated ZhongShan Hospital of Dalian University, Dalian, Liaoning, China
| | - Le Xiong
- Orthopaedic Department, Affiliated ZhongShan Hospital of Dalian University, Dalian, Liaoning, China
| | - Depeng Shang
- Orthopaedic Department, Affiliated ZhongShan Hospital of Dalian University, Dalian, Liaoning, China
| | - Yang Cong
- Orthopaedic Department, Affiliated ZhongShan Hospital of Dalian University, Dalian, Liaoning, China
| | - Dan Zhao
- Orthopaedic Department, Affiliated ZhongShan Hospital of Dalian University, Dalian, Liaoning, China
| | - Xiaowei Wei
- Orthopaedic Medical Research Center, Dalian University, Dalian, Liaoning, China
| | - Junlei Li
- Orthopaedic Medical Research Center, Dalian University, Dalian, Liaoning, China
| | - Dapeng Fu
- Orthopaedic Department, Affiliated ZhongShan Hospital of Dalian University, Dalian, Liaoning, China
| | - Haoyi Lian
- Orthopaedic Department, Affiliated ZhongShan Hospital of Dalian University, Dalian, Liaoning, China
| | - Zhenhua Zhao
- Orthopaedic Department, Affiliated ZhongShan Hospital of Dalian University, Dalian, Liaoning, China
| |
Collapse
|
2
|
Khazaeel K, Sadeghi A, Khademi Moghaddam F, Mohammadi T. The impact of graphene quantum dots on osteogenesis potential of Wharton's jelly mesenchymal stem cells in fibrin hydrogel scaffolds. Cytotechnology 2025; 77:14. [PMID: 39665046 PMCID: PMC11628478 DOI: 10.1007/s10616-024-00672-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 11/26/2024] [Indexed: 12/13/2024] Open
Abstract
Bone tissue engineering is a promising approach to overcome the limitations of traditional autograft bone transplantation. Graphene quantum dots (GQDs) have been suggested as an enhancement for osteogenic differentiation. This study aimed to investigate the ability of the fibrin hydrogel scaffold in the presence of graphene quantum dots to promote osteogenic differentiation of human Wharton's jelly-derived mesenchymal stem cells (hWJ-MSCs). The hWJ-MSCs were isolated from the Wharton's jelly of the human umbilical cord using a mechanical method. Fibrin hydrogel scaffolds were prepared by mixing 15 µl of thrombin solution with fibrinogen solution. GQDs were incorporated into the scaffolds at concentrations of 0, 5, and 10 µg/ml. Cell viability was determined through DAPI staining and the MTT assay. Osteogenic differentiation was assessed by measuring alkaline phosphatase (ALP) activity, quantifying calcium deposition using Alizarin Red S staining, and analyzing the gene expression of BGLAP, COL1A1, Runx-2 and ALP via qPCR. Scanning electron microscopy (SEM) was employed to analyze the scaffold architecture. SEM analysis revealed that the fibrin hydrogel exhibited a suitable architecture for tissue engineering, and DAPI staining confirmed cell viability. The MTT results indicated that the GQDs and fibrin hydrogel scaffold exhibited no cytotoxic effects. Furthermore, the incorporation of GQDs at a concentration of 10 µg/ml significantly enhanced ALP activity, calcium deposition, and the expression of osteogenesis-related genes compared to the control. The findings suggest that the combination of fibrin hydrogel and GQDs can effectively promote the osteogenic differentiation of hWJ-MSCs, contributing to the advancement of bone tissue engineering.
Collapse
Affiliation(s)
- Kaveh Khazaeel
- Department of Basic Sciences, Division of Anatomy and Embryology, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz, Iran
- Stem Cells and Transgenic Technology Research Center (STTRC), Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Abbas Sadeghi
- Department of Basic Sciences, Division of Anatomy and Embryology, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | | | - Tayebeh Mohammadi
- Department of Basic Sciences, Faculty of Veterinary Medicine, Razi University, Kermanshah, Iran
| |
Collapse
|
3
|
Agostinacchio F, Biada E, Gambari L, Grassi F, Bucciarelli A, Motta A. Surfactant-assisted photo-crosslinked silk fibroin sponges: A versatile platform for the design of bone scaffolds. BIOMATERIALS ADVANCES 2024; 161:213887. [PMID: 38735199 DOI: 10.1016/j.bioadv.2024.213887] [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: 10/23/2023] [Revised: 04/16/2024] [Accepted: 05/03/2024] [Indexed: 05/14/2024]
Abstract
Critical size bone defects cannot heal without aid and current clinical approaches exhibit some limitations, underling the need for novel solutions. Silk fibroin, derived from silkworms, is widely utilized in tissue engineering and regenerative medicine due to its remarkable properties, making it a promising candidate for bone tissue regeneration in vitro and in vivo. However, the clinical translation of silk-based materials requires refinements in 3D architecture, stability, and biomechanical properties. In earlier research, improved mechanical resistance and stability of chemically crosslinked methacrylate silk fibroin (Sil-Ma) sponges over physically crosslinked counterparts were highlighted. Furthermore, the influence of photo-initiator and surfactant concentrations on silk properties was investigated. However, the characterization of sponges with Sil-Ma solution concentrations above 10 % (w/V) was hindered by production optimization challenges, with only cell viability assessed. This study focuses on the evaluation of methacrylate sponges' suitability as temporal bone tissue regeneration scaffolds. Sil-Ma sponge fabrication at a fixed concentration of 20 % (w/V) was optimized and the impact of photo-initiator (LAP) concentrations and surfactant (Tween 80) presence/absence was studied. Their effects on pore formation, silk secondary structure, mechanical properties, and osteogenic differentiation of hBM-MSCs were investigated. We demonstrated that, by tuning silk sponges' composition, the optimal combination boosted osteogenic gene expression, offering a strategy to tailor biomechanical properties for effective bone regeneration. Utilizing Design of Experiment (DoE), correlations between sponge composition, porosity, and mechanical properties are established, guiding tailored material outcomes. Additionally, correlation matrices elucidate the microstructure's influence on gene expressions, providing insights for personalized approaches in bone tissue regeneration.
Collapse
Affiliation(s)
- Francesca Agostinacchio
- National Interuniversity Consortium of Material Science and Technology, Florence, Italy; BIOtech Research Center and European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Department of Industrial Engineering, University of Trento, Trento, Italy
| | - Elisa Biada
- BIOtech Research Center and European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Department of Industrial Engineering, University of Trento, Trento, Italy
| | - Laura Gambari
- Laboratorio Ramses, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Francesco Grassi
- Laboratorio Ramses, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | | | - Antonella Motta
- BIOtech Research Center and European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Department of Industrial Engineering, University of Trento, Trento, Italy.
| |
Collapse
|
4
|
Jankowski M, Farzaneh M, Ghaedrahmati F, Shirvaliloo M, Moalemnia A, Kulus M, Ziemak H, Chwarzyński M, Dzięgiel P, Zabel M, Piotrowska-Kempisty H, Bukowska D, Antosik P, Mozdziak P, Kempisty B. Unveiling Mesenchymal Stem Cells' Regenerative Potential in Clinical Applications: Insights in miRNA and lncRNA Implications. Cells 2023; 12:2559. [PMID: 37947637 PMCID: PMC10649218 DOI: 10.3390/cells12212559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 10/20/2023] [Accepted: 10/28/2023] [Indexed: 11/12/2023] Open
Abstract
It is now widely recognized that mesenchymal stem cells (MSCs) possess the capacity to differentiate into a wide array of cell types. Numerous studies have identified the role of lncRNA in the regulation of MSC differentiation. It is important to elucidate the role and interplay of microRNAs (miRNAs) and long non-coding RNAs (lncRNAs) in the regulation of signalling pathways that govern MSC function. Furthermore, miRNAs and lncRNAs are important clinical for innovative strategies aimed at addressing a wide spectrum of existing and emerging disease. Hence it is important to consider their impact on MSC function and differentiation. Examining the data available in public databases, we have collected the literature containing the latest discoveries pertaining to human stem cells and their potential in both fundamental research and clinical applications. Furthermore, we have compiled completed clinical studies that revolve around the application of MSCs, shedding light on the opportunities presented by harnessing the regulatory potential of miRNAs and lncRNAs. This exploration of the therapeutic possibilities offered by miRNAs and lncRNAs within MSCs unveils exciting prospects for the development of precision therapies and personalized treatment approaches. Ultimately, these advancements promise to augment the efficacy of regenerative strategies and produce positive outcomes for patients. As research in this field continues to evolve, it is imperative to explore and exploit the vast potential of miRNAs and lncRNAs as therapeutic agents. The findings provide a solid basis for ongoing investigations, fuelling the quest to fully unlock the regenerative potential of MSCs.
Collapse
Affiliation(s)
- Maurycy Jankowski
- Department of Computer Science and Statistics, Poznan University of Medical Sciences, 60-812 Poznan, Poland;
- Department of Histology and Embryology, Poznan University of Medical Sciences, 60-781 Poznan, Poland
| | - Maryam Farzaneh
- Fertility, Infertility and Perinatology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Farhoodeh Ghaedrahmati
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Milad Shirvaliloo
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Future Science Group, Unitec House, 2 Albert Place, London N3 1QB, UK
| | - Arash Moalemnia
- Faculty of Medicine, Dezful University of Medical Sciences, Dezful, Iran
| | - Magdalena Kulus
- Department of Veterinary Surgery, Institute of Veterinary Medicine, Nicolaus Copernicus University in Torun, 87-100 Torun, Poland
| | - Hanna Ziemak
- Department of Veterinary Surgery, Institute of Veterinary Medicine, Nicolaus Copernicus University in Torun, 87-100 Torun, Poland
| | - Mikołaj Chwarzyński
- Department of Veterinary Surgery, Institute of Veterinary Medicine, Nicolaus Copernicus University in Torun, 87-100 Torun, Poland
| | - Piotr Dzięgiel
- Division of Histology and Embryology, Department of Human Morphology and Embryology, Wroclaw Medical University, 50-368 Wroclaw, Poland
- Department of Physiotherapy, Wroclaw University School of Physical Education, 50-038 Wroclaw, Poland
| | - Maciej Zabel
- Division of Histology and Embryology, Department of Human Morphology and Embryology, Wroclaw Medical University, 50-368 Wroclaw, Poland
- Division of Anatomy and Histology, University of Zielona Góra, 65-046 Zielona Góra, Poland
| | - Hanna Piotrowska-Kempisty
- Department of Toxicology, Poznan University of Medical Sciences, 60-631 Poznan, Poland
- Department of Basic and Preclinical Sciences, Institute of Veterinary Medicine, Nicolaus Copernicus University in Torun, 87-100 Torun, Poland
| | - Dorota Bukowska
- Department of Diagnostics and Clinical Sciences, Institute of Veterinary Medicine, Nicolaus Copernicus University in Torun, 87-100 Torun, Poland
| | - Paweł Antosik
- Department of Veterinary Surgery, Institute of Veterinary Medicine, Nicolaus Copernicus University in Torun, 87-100 Torun, Poland
| | - Paul Mozdziak
- Prestage Department of Poultry Science, North Carolina State University, Raleigh, NC 27607, USA
- Physiology Graduate Faculty, North Carolina State University, Raleigh, NC 27613, USA
| | - Bartosz Kempisty
- Department of Veterinary Surgery, Institute of Veterinary Medicine, Nicolaus Copernicus University in Torun, 87-100 Torun, Poland
- Physiology Graduate Faculty, North Carolina State University, Raleigh, NC 27613, USA
- Division of Anatomy, Department of Human Morphology and Embryology, Wroclaw Medical University, 50-368 Wroclaw, Poland
- Department of Obstetrics and Gynecology, University Hospital and Masaryk University, 602 00 Brno, Czech Republic
| |
Collapse
|
5
|
Mousavi SJ, Ejeian F, Razmjou A, Nasr-Esfahani MH. In vivo evaluation of bone regeneration using ZIF8-modified polypropylene membrane in rat calvarium defects. J Clin Periodontol 2023; 50:1390-1405. [PMID: 37485621 DOI: 10.1111/jcpe.13855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2022] [Revised: 06/02/2023] [Accepted: 07/04/2023] [Indexed: 07/25/2023]
Abstract
AIM The profound potential of zeolitic imidazolate framework 8 (ZIF8) thin film for inducing osteogenesis has been previously established under in vitro conditions. As the next step towards the clinical application of ZIF8-modified substrates in periodontology, this in vivo study aimed to evaluate the ability of the ZIF8 crystalline layer to induce bone regeneration in an animal model defect. MATERIALS AND METHODS Following the mechanical characterization of the membranes and analysing the in vitro degradation of the ZIF8 layer, in vivo bone regeneration was evaluated in a critical-sized (5-mm) rat calvarial bone defect model. For each animal, one defect was randomly covered with either a polypropylene (PP) or a ZIF8-modified membrane (n = 7 per group), while the other defect was left untreated as a control. Eight weeks post surgery, bone formation was assessed by microcomputed tomography scanning, haematoxylin and eosin staining and immunohistochemical analysis. RESULTS The ZIF8-modified membrane outperformed the PP membrane in terms of mechanical properties and revealed a trace Zn+2 release. Results of in vivo evaluation verified the superior barrier function of the ZIF8-coated membrane compared with pristine PP membrane. Compared with the limited marginal bone formation in the control and PP groups, the defect area was almost filled with mature bone in the ZIF8-coated membrane group. CONCLUSIONS Our results support the effectiveness of the ZIF8-coated membrane as a promising material for improving clinical outcomes of guided bone regeneration procedures, without using biological components.
Collapse
Affiliation(s)
- Seyed Javad Mousavi
- Department of Biotechnology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran
- Department of Animal Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Fatemeh Ejeian
- Department of Animal Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Amir Razmjou
- School of Engineering, Edith Cowan University, Perth, Western Australia, Australia
- UNESCO Centre for Membrane Science and Technology, School of Chemical Engineering, University of New South Wales, Sydney, New South Wales, Australia
| | - Mohammad Hossein Nasr-Esfahani
- Department of Animal Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| |
Collapse
|
6
|
Rifai A, Weerasinghe DK, Tilaye GA, Nisbet D, Hodge JM, Pasco JA, Williams LJ, Samarasinghe RM, Williams RJ. Biofabrication of functional bone tissue: defining tissue-engineered scaffolds from nature. Front Bioeng Biotechnol 2023; 11:1185841. [PMID: 37614632 PMCID: PMC10444209 DOI: 10.3389/fbioe.2023.1185841] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 07/24/2023] [Indexed: 08/25/2023] Open
Abstract
Damage to bone leads to pain and loss of movement in the musculoskeletal system. Although bone can regenerate, sometimes it is damaged beyond its innate capacity. Research interest is increasingly turning to tissue engineering (TE) processes to provide a clinical solution for bone defects. Despite the increasing biomimicry of tissue-engineered scaffolds, significant gaps remain in creating the complex bone substitutes, which include the biochemical and physical conditions required to recapitulate bone cells' natural growth, differentiation and maturation. Combining advanced biomaterials with new additive manufacturing technologies allows the development of 3D tissue, capable of forming cell aggregates and organoids based on natural and stimulated cues. Here, we provide an overview of the structure and mechanical properties of natural bone, the role of bone cells, the remodelling process, cytokines and signalling pathways, causes of bone defects and typical treatments and new TE strategies. We highlight processes of selecting biomaterials, cells and growth factors. Finally, we discuss innovative tissue-engineered models that have physiological and anatomical relevance for cancer treatments, injectable stimuli gels, and other therapeutic drug delivery systems. We also review current challenges and prospects of bone TE. Overall, this review serves as guide to understand and develop better tissue-engineered bone designs.
Collapse
Affiliation(s)
- Aaqil Rifai
- Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, VIC, Australia
| | - D. Kavindi Weerasinghe
- Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, VIC, Australia
| | - Gebreselassie Addisu Tilaye
- Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, VIC, Australia
| | - David Nisbet
- The Graeme Clark Institute, The University of Melbourne, Melbourne, VIC, Australia
- Department of Biomedical Engineering, Faculty of Engineering and Information Technology, The University of Melbourne, Melbourne, VIC, Australia
- Melbourne Medical School, Faculty of Medicine, Dentistry and Health Science, The University of Melbourne, Melbourne, VIC, Australia
- Laboratory of Advanced Biomaterials, John Curtin School of Medical Research, Australian National University, Canberra, ACT, Australia
- Aikenhead Centre for Medical Discovery, St. Vincent’s Hospital, Melbourne, VIC, Australia
| | - Jason M. Hodge
- Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, VIC, Australia
- Barwon Health, Geelong, VIC, Australia
| | - Julie A. Pasco
- Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, VIC, Australia
- Barwon Health, Geelong, VIC, Australia
- Department of Epidemiology and Preventive Medicine, Monash University, Melbourne, VIC, Australia
- Department of Medicine-Western Health, The University of Melbourne, St Albans, VIC, Australia
| | - Lana J. Williams
- Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, VIC, Australia
- Barwon Health, Geelong, VIC, Australia
| | - Rasika M. Samarasinghe
- Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, VIC, Australia
| | - Richard J. Williams
- Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, VIC, Australia
- The Graeme Clark Institute, The University of Melbourne, Melbourne, VIC, Australia
- Aikenhead Centre for Medical Discovery, St. Vincent’s Hospital, Melbourne, VIC, Australia
| |
Collapse
|
7
|
Zhang Z, Bi F, Guo W. Research Advances on Hydrogel-Based Materials for Tissue Regeneration and Remineralization in Tooth. Gels 2023; 9:gels9030245. [PMID: 36975694 PMCID: PMC10048036 DOI: 10.3390/gels9030245] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Revised: 03/14/2023] [Accepted: 03/17/2023] [Indexed: 03/29/2023] Open
Abstract
Tissue regeneration and remineralization in teeth is a long-term and complex biological process, including the regeneration of pulp and periodontal tissue, and re-mineralization of dentin, cementum and enamel. Suitable materials are needed to provide cell scaffolds, drug carriers or mineralization in this environment. These materials need to regulate the unique odontogenesis process. Hydrogel-based materials are considered good scaffolds for pulp and periodontal tissue repair in the field of tissue engineering due to their inherent biocompatibility and biodegradability, slow release of drugs, simulation of extracellular matrix, and the ability to provide a mineralized template. The excellent properties of hydrogels make them particularly attractive in the research of tissue regeneration and remineralization in teeth. This paper introduces the latest progress of hydrogel-based materials in pulp and periodontal tissue regeneration and hard tissue mineralization and puts forward prospects for their future application. Overall, this review reveals the application of hydrogel-based materials in tissue regeneration and remineralization in teeth.
Collapse
Affiliation(s)
- Zhijun Zhang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
- National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
- Department of Pediatric Dentistry, West China School of Stomatology, Sichuan University, Chengdu 610041, China
| | - Fei Bi
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
- National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Weihua Guo
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
- National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
- Department of Pediatric Dentistry, West China School of Stomatology, Sichuan University, Chengdu 610041, China
- Yunnan Key Laboratory of Stomatology, The Affiliated Hospital of Stomatology, School of Stomatology, Kunming Medical University, Kunming 650500, China
| |
Collapse
|
8
|
Nadine S, Fernandes I, Patrício SG, Correia CR, Mano JF. Liquefied Microcapsules Compartmentalizing Macrophages and Umbilical Cord-Derived Cells for Bone Tissue Engineering. Adv Healthc Mater 2022; 11:e2200651. [PMID: 35904030 DOI: 10.1002/adhm.202200651] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 07/22/2022] [Indexed: 01/28/2023]
Abstract
Extraordinary capabilities underlie the potential use of immune cells, particularly macrophages, in bone tissue engineering. Indeed, the depletion of macrophages during bone repair often culminates in disease scenarios. Inspired by the native dynamics between immune and skeletal systems, this work proposes a straightforward in vitro method to bioengineer biomimetic bone niches using biological waste. For that, liquefied and semipermeable reservoirs generated by electrohydrodynamic atomization and layer-by-layer techniques are developed to coculture umbilical cord-derived human cells, namely monocyte-derived macrophages, mesenchymal-derived stromal cells (MSCs), and human umbilical vein endothelial cells (HUVECs). Poly(ε-caprolactone) microparticles are also added to the liquefied core to act as cell carriers. The fabricated microcapsules grant the successful development of viable microtissues, ensuring the high diffusion of bioactive factors. Interestingly, macrophages within the bioengineered microcapsules increase the release of osteocalcin, osteoprotegerin, and vascular endothelial growth factor. The cytokines profile variation indicates macrophages' polarization into a prohealing phenotype. Altogether, the incorporation of macrophages within the fabricated microcapsules allows to recreate an appropriate bone microenvironment for developing new bone mineralized microtissues. The proposed bioencapsulation protocol is a powerful self-regulated system, which might find great applicability in bone tissue engineering based on bottom-up approaches or disease modeling.
Collapse
Affiliation(s)
- Sara Nadine
- CICECO - Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, Campus Universitário de Santiago, Aveiro, 3810-193, Portugal
| | - Inês Fernandes
- CICECO - Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, Campus Universitário de Santiago, Aveiro, 3810-193, Portugal
| | - Sónia G Patrício
- CICECO - Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, Campus Universitário de Santiago, Aveiro, 3810-193, Portugal
| | - Clara R Correia
- CICECO - Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, Campus Universitário de Santiago, Aveiro, 3810-193, Portugal
| | - João F Mano
- CICECO - Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, Campus Universitário de Santiago, Aveiro, 3810-193, Portugal
| |
Collapse
|
9
|
Costa M, Lima R, Alves N, Silva N, Gasik M, Silva F, Bartolomeu F, Miranda G. Multi-material cellular structured orthopedic implants design: In vitro and bio-tribological performance. J Mech Behav Biomed Mater 2022; 131:105246. [DOI: 10.1016/j.jmbbm.2022.105246] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 04/11/2022] [Accepted: 04/17/2022] [Indexed: 12/15/2022]
|
10
|
Hurtado A, Aljabali AAA, Mishra V, Tambuwala MM, Serrano-Aroca Á. Alginate: Enhancement Strategies for Advanced Applications. Int J Mol Sci 2022; 23:4486. [PMID: 35562876 PMCID: PMC9102972 DOI: 10.3390/ijms23094486] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Revised: 04/14/2022] [Accepted: 04/17/2022] [Indexed: 02/06/2023] Open
Abstract
Alginate is an excellent biodegradable and renewable material that is already used for a broad range of industrial applications, including advanced fields, such as biomedicine and bioengineering, due to its excellent biodegradable and biocompatible properties. This biopolymer can be produced from brown algae or a microorganism culture. This review presents the principles, chemical structures, gelation properties, chemical interactions, production, sterilization, purification, types, and alginate-based hydrogels developed so far. We present all of the advanced strategies used to remarkably enhance this biopolymer's physicochemical and biological characteristics in various forms, such as injectable gels, fibers, films, hydrogels, and scaffolds. Thus, we present here all of the material engineering enhancement approaches achieved so far in this biopolymer in terms of mechanical reinforcement, thermal and electrical performance, wettability, water sorption and diffusion, antimicrobial activity, in vivo and in vitro biological behavior, including toxicity, cell adhesion, proliferation, and differentiation, immunological response, biodegradation, porosity, and its use as scaffolds for tissue engineering applications. These improvements to overcome the drawbacks of the alginate biopolymer could exponentially increase the significant number of alginate applications that go from the paper industry to the bioprinting of organs.
Collapse
Affiliation(s)
- Alejandro Hurtado
- Biomaterials and Bioengineering Laboratory, Centro de Investigación Traslacional San Alberto Magno, Universidad Católica de Valencia San Vicente Mártir, c/Guillem de Castro 94, 46001 Valencia, Spain;
| | - Alaa A. A. Aljabali
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Yarmouk University, Irbid 21163, Jordan;
| | - Vijay Mishra
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara 144411, Punjab, India;
| | - Murtaza M. Tambuwala
- School of Pharmacy and Pharmaceutical Science, Ulster University, Coleraine BT52 1SA, Northern Ireland, UK;
| | - Ángel Serrano-Aroca
- Biomaterials and Bioengineering Laboratory, Centro de Investigación Traslacional San Alberto Magno, Universidad Católica de Valencia San Vicente Mártir, c/Guillem de Castro 94, 46001 Valencia, Spain;
| |
Collapse
|
11
|
Maleki-Ghaleh H, Siadati MH, Omidi Y, Kavanlouei M, Barar J, Akbari-Fakhrabadi A, Adibkia K, Beygi-Khosrowshahi Y. Synchrotron SAXS/WAXS and TEM studies of zinc doped natural hydroxyapatite nanoparticles and their evaluation on osteogenic differentiation of human mesenchymal stem cells. MATERIALS CHEMISTRY AND PHYSICS 2022; 276:125346. [DOI: 10.1016/j.matchemphys.2021.125346] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
12
|
Microfluidic-templating alginate microgels crosslinked by different metal ions as engineered microenvironment to regulate stem cell behavior for osteogenesis. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 131:112497. [PMID: 34857283 DOI: 10.1016/j.msec.2021.112497] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 10/08/2021] [Accepted: 10/13/2021] [Indexed: 12/28/2022]
Abstract
Cell microenvironment is a collection of dynamic biochemical and biophysical cues which functions as the key factor in determining cell behavior. Encapsulating single cell into micrometer-scale hydrogels which mimics the cell microenvironment can be used for single cell analysis, cell therapies, and tissue engineering. Here, we developed a microfluidics-based platform to engineer the niche environment at single cell level using alginate microgels crosslinked by different metal ions to regulate stem cell behavior for bone regeneration. Specifically, we revealed that Ca2+ in the engineered microenvironment promoted osteogenic differentiation of encapsulated stem cells and substantially accelerated the matrix mineralization compared to Sr2+in vitro. However, the superior osteoinductive capacity of Ca2+ compared with Sr2+ led to comparable bone healing in a rat bone defect model. This attributed to Sr2+ in microgels to inhibit the osteoclast activity and bone resorption after implantation. In summary, the present study demonstrates metal ions as a critical factor in the environmental cues to affect cell behavior and influence the efficacy of stem cell-based therapy in tissue regeneration, and provides new insights to engineer an expecting microenvironment for regenerative medicine.
Collapse
|
13
|
Kaibara T, Wang L, Tsuda M, Nonoyama T, Kurokawa T, Iwasaki N, Gong JP, Tanaka S, Yasuda K. Hydroxyapatite-hybridized double-network hydrogel surface enhances differentiation of bone marrow-derived mesenchymal stem cells to osteogenic cells. J Biomed Mater Res A 2021; 110:747-760. [PMID: 34713570 DOI: 10.1002/jbm.a.37324] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 09/22/2021] [Accepted: 10/15/2021] [Indexed: 01/31/2023]
Abstract
Recently, we have developed a hydroxyapatite (HAp)-hybridized double-network (DN) hydrogel (HAp/DN gel), which can robustly bond to the bone tissue in the living body. The purpose of this study is to clarify whether the HAp/DN gel surface can differentiate the bone marrow-derived mesenchymal stem cells (MSCs) to osteogenic cells. We used the MSCs which were harvested from the rabbit bone marrow and cultured on the polystyrene (PS) dish using the autogenous serum-supplemented medium. First, we confirmed the properties of MSCs by evaluating colony forming unit capacity, expression of MSC markers using flow cytometry, and multidifferential capacity. Secondly, polymerase chain reaction analysis demonstrated that the HAp/DN gel surface significantly enhanced mRNA expression of the eight osteogenic markers (TGF-β1, BMP-2, Runx2, Col-1, ALP, OPN, BSP, and OCN) in the cultured MSCs at 7 days than the PS surfaces (p < 0.0001), while the DN gel and HAp surfaces provided no or only a slight effect on the expression of these markers except for Runx2. Additionally, the alkaline phosphatase activity was significantly higher in the cells cultured on the HAp/DN gel surface than in the other three material surfaces (p < 0.0001). Thirdly, when the HAp/DN gel plug was implanted into the rabbit bone marrow, MSC marker-positive cells were recruited in the tissue generated around the plug at 3 days, and Runx2 and OCN were highly expressed in these cells. In conclusion, this study demonstrated that the HAp/DN gel surface can differentiate the MSCs into osteogenic cells.
Collapse
Affiliation(s)
- Takuma Kaibara
- Department of Orthopaedic Surgery, Hokkaido University Graduate School of Medicine, Sapporo, Japan.,Department of Cancer Pathology, Faculty of Medicine, Hokkaido University, Sapporo, Japan
| | - Lei Wang
- Department of Cancer Pathology, Faculty of Medicine, Hokkaido University, Sapporo, Japan.,Global Station for Soft Matter, Global Institution for Collaborative Research and Education (GI-CoRE), Hokkaido University, Sapporo, Japan
| | - Masumi Tsuda
- Department of Cancer Pathology, Faculty of Medicine, Hokkaido University, Sapporo, Japan.,Global Station for Soft Matter, Global Institution for Collaborative Research and Education (GI-CoRE), Hokkaido University, Sapporo, Japan.,Institute for Chemical Reaction Design and Discovery (WPI-ICReDD), Hokkaido University, Sapporo, Japan
| | - Takayuki Nonoyama
- Global Station for Soft Matter, Global Institution for Collaborative Research and Education (GI-CoRE), Hokkaido University, Sapporo, Japan.,Laboratory of Soft & Wet Matter, Faculty of Advanced Life Science, Hokkaido University, Sapporo, Japan
| | - Takayuki Kurokawa
- Global Station for Soft Matter, Global Institution for Collaborative Research and Education (GI-CoRE), Hokkaido University, Sapporo, Japan.,Laboratory of Soft & Wet Matter, Faculty of Advanced Life Science, Hokkaido University, Sapporo, Japan
| | - Norimasa Iwasaki
- Department of Orthopaedic Surgery, Hokkaido University Graduate School of Medicine, Sapporo, Japan.,Global Station for Soft Matter, Global Institution for Collaborative Research and Education (GI-CoRE), Hokkaido University, Sapporo, Japan
| | - Jian Ping Gong
- Global Station for Soft Matter, Global Institution for Collaborative Research and Education (GI-CoRE), Hokkaido University, Sapporo, Japan.,Institute for Chemical Reaction Design and Discovery (WPI-ICReDD), Hokkaido University, Sapporo, Japan.,Laboratory of Soft & Wet Matter, Faculty of Advanced Life Science, Hokkaido University, Sapporo, Japan
| | - Shinya Tanaka
- Department of Cancer Pathology, Faculty of Medicine, Hokkaido University, Sapporo, Japan.,Global Station for Soft Matter, Global Institution for Collaborative Research and Education (GI-CoRE), Hokkaido University, Sapporo, Japan.,Institute for Chemical Reaction Design and Discovery (WPI-ICReDD), Hokkaido University, Sapporo, Japan
| | - Kazunori Yasuda
- Global Station for Soft Matter, Global Institution for Collaborative Research and Education (GI-CoRE), Hokkaido University, Sapporo, Japan.,Sports Medicine and Arthroscopy Center, Yagi Orthopaedic Hospital, Sapporo, Japan
| |
Collapse
|
14
|
Xu S, Xie X, Li C, Liu Z, Zuo D. Micromolar sodium fluoride promotes osteo/odontogenic differentiation in dental pulp stem cells by inhibiting PI3K/AKT pathway. Arch Oral Biol 2021; 131:105265. [PMID: 34601318 DOI: 10.1016/j.archoralbio.2021.105265] [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: 07/12/2021] [Revised: 08/19/2021] [Accepted: 09/12/2021] [Indexed: 12/19/2022]
Abstract
OBJECTIVE Sodium fluoride (NaF) plays an important role in preventing dental caries. However, the regulatory effect of NaF on the committed differentiation of DPSCs is not fully understood. In this study, we characterized the impact of micromolar levels of NaF on the osteo/odontogenic differentiation of DPSCs. DESIGN DPSCs were isolated from healthy human third molars and were cultured in conditioned media with different concentrations of NaF. RNA sequencing (RNA-seq) combined with Kyoto Encyclopedia of Genes and Genomes pathway enrichment analysis was used to assess the pathways regulated by NaF. Alkaline phosphatase activity, Alizarin red staining, Western blotting, and real-time qRT-PCR were used to determine the osteo/odontogenic differentiation in DPSCs treated with NaF. RESULTS NaF significantly promoted the osteo/odontogenic differentiation of DPSCs at micromolar levels. Furthermore, RNA-seq and KEGG pathway enrichment analysis indicated that the PI3K/AKT pathway was involved in the pro-osteoclastogenesis effect of NaF. Western blotting analysis exhibited that the phosphorylation of AKT was decreased in NaF-treated DPSCs. Chemical inhibition of the PI3K/AKT pathway abrogated the NaF-promoted DPSCs osteo/odontogenic differentiation. CONCLUSION Micromolar NaF can promote the osteo/odontogenic differentiation of DPSCs by inhibiting the PI3K/AKT pathway. DATA AVAILABILITY The data used to support the findings of this study are available from the corresponding author upon request.
Collapse
Affiliation(s)
- Shuaimei Xu
- Department of Endodontics, Stomatological Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China; Department of Medical Laboratory, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Xinghuan Xie
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Changzhou Li
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Zhongjun Liu
- Department of Endodontics, Stomatological Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Daming Zuo
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong 510515, China.
| |
Collapse
|
15
|
Suryani L, Foo JKR, Cardilla A, Dong Y, Muthukumaran P, Hassanbhai A, Wen F, Simon DT, Iandolo D, Yu N, Ng KW, Teoh SH. Effects of Pulsed Electromagnetic Field Intensity on Mesenchymal Stem Cells. Bioelectricity 2021. [DOI: 10.1089/bioe.2021.0002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Luvita Suryani
- School of Chemical and Biomedical Engineering, College of Engineering, Nanyang Technological University, Singapore, Singapore
| | - Jyong Kiat Reuben Foo
- School of Chemical and Biomedical Engineering, College of Engineering, Nanyang Technological University, Singapore, Singapore
| | - Angelysia Cardilla
- School of Chemical and Biomedical Engineering, College of Engineering, Nanyang Technological University, Singapore, Singapore
| | - Yibing Dong
- School of Materials Science and Engineering, Nanyang Technological University, Singapore, Singapore
| | - Padmalosini Muthukumaran
- School of Chemical and Biomedical Engineering, College of Engineering, Nanyang Technological University, Singapore, Singapore
| | - Ammar Hassanbhai
- School of Chemical and Biomedical Engineering, College of Engineering, Nanyang Technological University, Singapore, Singapore
| | - Feng Wen
- School of Chemical and Biomedical Engineering, College of Engineering, Nanyang Technological University, Singapore, Singapore
| | - Daniel T. Simon
- Laboratory of Organic Electronics, Department of Science and Technology, Linköping University, Norrköping, Sweden
| | - Donata Iandolo
- Laboratory of Organic Electronics, Department of Science and Technology, Linköping University, Norrköping, Sweden
- UMR5510 MATEIS, CNRS, INSA-Lyon, University of Lyon, Lyon, France
- Mines Saint-Etienne, INSERM, U1059 SAINBIOSE, Saint-Étienne, France
| | - Na Yu
- National Dental Centre Singapore, Singapore, Singapore
- Duke-NUS Medical School, Singapore, Singapore
| | - Kee Woei Ng
- School of Materials Science and Engineering, Nanyang Technological University, Singapore, Singapore
- Environmental Chemistry & Materials Centre, Nanyang Environment and Water Research Institute (NEWRI), Nanyang Technological University, Singapore, Singapore
- Center for Nanotechnology and Nanotoxicology, Harvard T.H. Chan School of Public Health, Harvard University, Boston, Massachusetts, USA
| | - Swee-Hin Teoh
- School of Chemical and Biomedical Engineering, College of Engineering, Nanyang Technological University, Singapore, Singapore
- Lee Kong Chian School of Medicine, Singapore, Singapore
| |
Collapse
|
16
|
Ødegaard KS, Ouyang L, Ma Q, Buene G, Wan D, Elverum CW, Torgersen J, Standal T, Westhrin M. Revealing the influence of electron beam melted Ti-6Al-4V scaffolds on osteogenesis of human bone marrow-derived mesenchymal stromal cells. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2021; 32:97. [PMID: 34406475 PMCID: PMC8373740 DOI: 10.1007/s10856-021-06572-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 07/23/2021] [Indexed: 06/13/2023]
Abstract
Porous Titanium-6Aluminum-4Vanadium scaffolds made by electron beam-based additive manufacturing (AM) have emerged as state-of-the-art implant devices. However, there is still limited knowledge on how they influence the osteogenic differentiation of bone marrow-derived mesenchymal stromal cells (BMSCs). In this study, BMSCs are cultured on such porous scaffolds to determine how the scaffolds influence the osteogenic differentiation of the cells. The scaffolds are biocompatible, as revealed by the increasing cell viability. Cells are evenly distributed on the scaffolds after 3 days of culturing followed by an increase in bone matrix development after 21 days of culturing. qPCR analysis provides insight into the cells' osteogenic differentiation, where RUNX2 expression indicate the onset of differentiation towards osteoblasts. The COL1A1 expression suggests that the differentiated osteoblasts can produce the osteoid. Alkaline phosphatase staining indicates an onset of mineralization at day 7 in OM. The even deposits of calcium at day 21 further supports a successful bone mineralization. This work shines light on the interplay between AM Ti64 scaffolds and bone growth, which may ultimately lead to a new way of creating long lasting bone implants with fast recovery times.
Collapse
Affiliation(s)
- Kristin S Ødegaard
- Department of Mechanical and Industrial Engineering, Norwegian University of Science and Technology, Trondheim, Norway
| | - Lingzi Ouyang
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Qianli Ma
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Glenn Buene
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Di Wan
- Department of Mechanical and Industrial Engineering, Norwegian University of Science and Technology, Trondheim, Norway
| | - Christer W Elverum
- Department of Mechanical and Industrial Engineering, Norwegian University of Science and Technology, Trondheim, Norway
| | - Jan Torgersen
- Department of Mechanical and Industrial Engineering, Norwegian University of Science and Technology, Trondheim, Norway.
| | - Therese Standal
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Marita Westhrin
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| |
Collapse
|
17
|
Yang Y, Liu S, He C, Chen Z, Lyu T, Zeng L, Wang L, Zhang F, Chen H, Zhao RC. Long Non-coding RNA Regulation of Mesenchymal Stem Cell Homeostasis and Differentiation: Advances, Challenges, and Perspectives. Front Cell Dev Biol 2021; 9:711005. [PMID: 34368161 PMCID: PMC8339964 DOI: 10.3389/fcell.2021.711005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 06/21/2021] [Indexed: 12/25/2022] Open
Abstract
Given the self-renewal, multi-differentiation, immunoregulatory, and tissue maintenance properties, mesenchymal stem cells (MSCs) are promising candidates for stem cell-based therapies. Breakthroughs have been made in uncovering MSCs as key contributors to homeostasis and the regenerative repair of tissues and organs derived from three germ layers. MSC differentiation into specialized cell types is sophisticatedly regulated, and accumulating evidence suggests long non-coding RNAs (lncRNAs) as the master regulators of various biological processes including the maintenance of homeostasis and multi-differentiation functions through epigenetic, transcriptional, and post-translational mechanisms. LncRNAs are ubiquitous and generally referred to as non-coding transcripts longer than 200 bp. Most lncRNAs are evolutionary conserved and species-specific; however, the weak conservation of their sequences across species does not affect their diverse biological functions. Although numerous lncRNAs have been annotated and studied, they are nevertheless only the tip of the iceberg; the rest remain to be discovered. In this review, we characterize MSC functions in homeostasis and highlight recent advances on the functions and mechanisms of lncRNAs in regulating MSC homeostasis and differentiation. We also discuss the current challenges and perspectives for understanding the roles of lncRNAs in MSC functions in homeostasis, which could help develop promising targets for MSC-based therapies.
Collapse
Affiliation(s)
- Yanlei Yang
- Key Laboratory of the Ministry of Education, Department of Rheumatology and Clinical Immunology, Clinical Immunology Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China.,Beijing Key Laboratory (No. BZO381), School of Basic Medicine, Center of Excellence in Tissue Engineering, Peking Union Medical College Hospital, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Suying Liu
- Key Laboratory of the Ministry of Education, Department of Rheumatology and Clinical Immunology, Clinical Immunology Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Chengmei He
- Key Laboratory of the Ministry of Education, Department of Rheumatology and Clinical Immunology, Clinical Immunology Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Zhilei Chen
- Key Laboratory of the Ministry of Education, Department of Rheumatology and Clinical Immunology, Clinical Immunology Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Taibiao Lyu
- Key Laboratory of the Ministry of Education, Department of Rheumatology and Clinical Immunology, Clinical Immunology Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Liuting Zeng
- Key Laboratory of the Ministry of Education, Department of Rheumatology and Clinical Immunology, Clinical Immunology Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Li Wang
- Key Laboratory of the Ministry of Education, Department of Rheumatology and Clinical Immunology, Clinical Immunology Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Fengchun Zhang
- Key Laboratory of the Ministry of Education, Department of Rheumatology and Clinical Immunology, Clinical Immunology Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Hua Chen
- Key Laboratory of the Ministry of Education, Department of Rheumatology and Clinical Immunology, Clinical Immunology Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Robert Chunhua Zhao
- Beijing Key Laboratory (No. BZO381), School of Basic Medicine, Center of Excellence in Tissue Engineering, Peking Union Medical College Hospital, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China.,School of Life Sciences, Shanghai University, Shanghai, China
| |
Collapse
|
18
|
Kulanthaivel S, Agarwal T, Sharan Rathnam VS, Pal K, Banerjee I. Cobalt doped nano-hydroxyapatite incorporated gum tragacanth-alginate beads as angiogenic-osteogenic cell encapsulation system for mesenchymal stem cell based bone tissue engineering. Int J Biol Macromol 2021; 179:101-115. [PMID: 33621571 DOI: 10.1016/j.ijbiomac.2021.02.136] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 02/10/2021] [Accepted: 02/18/2021] [Indexed: 12/14/2022]
Abstract
Angiogenic-osteogenic cell encapsulation system is a technical need for human mesenchymal stem cell (hMSC)-based bone tissue engineering (BTE). Here, we have developed a highly efficient hMSC encapsulation system by incorporating bivalent cobalt doped nano-hydroxyapatite (HAN) and gum tragacanth (GT) as angiogenic-osteogenic components into the calcium alginate (CA) beads. Physico-chemical characterizations revealed that the swelling and degradation of HAN incorporated CA-GT beads (GT-HAN) were 1.34 folds and 2 folds higher than calcium alginate (CA) beads. Furthermore, the diffusion coefficient of solute molecule was found 2.5-fold higher in GT-HAN with respect to CA bead. It is observed that GT-HAN supports the long-term viability of encapsulated hMSC and causes 50% less production of reactive oxygen species (ROS) in comparison to CA beads. The expression of osteogenic differentiation markers was found 1.5-2.5 folds higher in the case of GT-HAN in comparison to CA. A similar trend was observed for hypoxia inducible factor 1 alpha (HIF-1α) and vascular endothelial growth factor (VEGF). The soluble secretome from hMSC encapsulated in the GT-HAN induced proliferation of endothelial cells and supported tube formation (2.5-fold higher than CA beads). These results corroborated that GT-HAN could be used as an angiogenic-osteogenic cell encapsulation matrix for hMSC encapsulation and BTE application.
Collapse
Affiliation(s)
- Senthilguru Kulanthaivel
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology Delhi, New Delhi 110016, India
| | - Tarun Agarwal
- Department of Biotechnology, Indian Institute of Technology Kharagpur, West Bengal 721302, India
| | - V S Sharan Rathnam
- Department of Biotechnology and Medical Engineering, National Institute of Technology, Rourkela, Odisha 769008, India
| | - Kunal Pal
- Department of Biotechnology and Medical Engineering, National Institute of Technology, Rourkela, Odisha 769008, India
| | - Indranil Banerjee
- Department of Bioscience and Bioengineering, Indian Institute of Technology Jodhpur, Rajasthan 342037, India.
| |
Collapse
|
19
|
Prouvé E, Drouin B, Chevallier P, Rémy M, Durrieu MC, Laroche G. Evaluating Poly(Acrylamide-co-Acrylic Acid) Hydrogels Stress Relaxation to Direct the Osteogenic Differentiation of Mesenchymal Stem Cells. Macromol Biosci 2021; 21:e2100069. [PMID: 33870650 DOI: 10.1002/mabi.202100069] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 03/30/2021] [Indexed: 11/09/2022]
Abstract
The aim of this study is to investigate polyacrylamide-based hydrogels stress relaxation and the subsequent impact on the osteogenic differentiation of human mesenchymal stem cells (hMSCs). Different hydrogels are synthesized by varying the amount of cross-linker and the ratio between the monomers (acrylamide and acrylic acid), and characterized by compression tests. It has been found that hydrogels containing 18% of acrylic acid exhibit an average relaxation of 70%, while pure polyacrylamide gels show an average relaxation of 15%. Subsequently, hMSCs are cultured on two different hydrogels functionalized with a mimetic peptide of the bone morphogenetic protein-2 to enable cell adhesion and favor their osteogenic differentiation. Phalloidin staining shows that for a constant stiffness of 55 kPa, a hydrogel with a low relaxation (15%) leads to star-shaped cells, which is typical of osteocytes, while a hydrogel with a high relaxation (70%) presents cells with a polygonal shape characteristic of osteoblasts. Immunofluorescence labeling of E11, strongly expressed in early osteocytes, also shows a dramatically higher expression for cells cultured on the hydrogel with low relaxation (15%). These results clearly demonstrate that, by fine-tuning hydrogels stress relaxation, hMSCs differentiation can be directed toward osteoblasts, and even osteocytes, which is particularly rare in vitro.
Collapse
Affiliation(s)
- Emilie Prouvé
- Department of mining, metallurgy, and materials engineering, Surface Engineering Laboratory, Research Center on Advanced Materials, Laval University, 1065 Avenue de la médecine, Québec, G1V 0A6, Canada.,Research Center of the University Hospital of Québec, Regenerative Medicine axis, St-François d'Assise Hospital, Laval University, 10 rue de l'Espinay, Québec, G1L 3L5, Canada.,Institute of Chemistry and Biology of Membranes and Nano-objects (UMR 5248 CBMN), Bordeaux University, Allée Geoffroy St Hilaire - Bât B14, Pessac, 33600, France.,CNRS, CBMN UMR5248, Allée Geoffroy Saint Hilaire - Bât B14, Pessac, 33600, France.,Bordeaux INP, CBMN UMR5248, Allée Geoffroy Saint Hilaire - Bât B14, Pessac, 33600, France
| | - Bernard Drouin
- Research Center of the University Hospital of Québec, Regenerative Medicine axis, St-François d'Assise Hospital, Laval University, 10 rue de l'Espinay, Québec, G1L 3L5, Canada
| | - Pascale Chevallier
- Department of mining, metallurgy, and materials engineering, Surface Engineering Laboratory, Research Center on Advanced Materials, Laval University, 1065 Avenue de la médecine, Québec, G1V 0A6, Canada.,Research Center of the University Hospital of Québec, Regenerative Medicine axis, St-François d'Assise Hospital, Laval University, 10 rue de l'Espinay, Québec, G1L 3L5, Canada
| | - Murielle Rémy
- Institute of Chemistry and Biology of Membranes and Nano-objects (UMR 5248 CBMN), Bordeaux University, Allée Geoffroy St Hilaire - Bât B14, Pessac, 33600, France.,CNRS, CBMN UMR5248, Allée Geoffroy Saint Hilaire - Bât B14, Pessac, 33600, France.,Bordeaux INP, CBMN UMR5248, Allée Geoffroy Saint Hilaire - Bât B14, Pessac, 33600, France
| | - Marie-Christine Durrieu
- Institute of Chemistry and Biology of Membranes and Nano-objects (UMR 5248 CBMN), Bordeaux University, Allée Geoffroy St Hilaire - Bât B14, Pessac, 33600, France.,CNRS, CBMN UMR5248, Allée Geoffroy Saint Hilaire - Bât B14, Pessac, 33600, France.,Bordeaux INP, CBMN UMR5248, Allée Geoffroy Saint Hilaire - Bât B14, Pessac, 33600, France
| | - Gaétan Laroche
- Department of mining, metallurgy, and materials engineering, Surface Engineering Laboratory, Research Center on Advanced Materials, Laval University, 1065 Avenue de la médecine, Québec, G1V 0A6, Canada.,Research Center of the University Hospital of Québec, Regenerative Medicine axis, St-François d'Assise Hospital, Laval University, 10 rue de l'Espinay, Québec, G1L 3L5, Canada
| |
Collapse
|
20
|
Osteogenic differentiation of hBMSCs on porous photo-crosslinked poly(trimethylene carbonate) and nano-hydroxyapatite composites. Eur Polym J 2021. [DOI: 10.1016/j.eurpolymj.2021.110335] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
21
|
Tejo-Otero A, Ritchie AC. Biological and mechanical evaluation of mineralized-hydrogel scaffolds for tissue engineering applications. J Biomater Appl 2021; 36:460-473. [PMID: 33596707 DOI: 10.1177/0885328221995425] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Chitosan and gelatin have been extensively used in tissue engineering for a wide range of different applications, such as wound healing or bone regeneration, due to their advantages: excellent biocompatibility (promoting cell adhesion and proliferation), low price and biodegradability. Nonetheless, their main drawback is that they have poor mechanical properties, consequently restricting their use in bone tissue engineering. In previous studies, both materials were cross-linked, with added calcium minerals, which led to an improvement in both mechanical and biological properties. Therefore, this study carries out a mechanical and biological characterization of mineral-hydrogel scaffolds in order to find the best compositions. Different proportions of calcium compounds (CaCO3 and CaHPO4) are used to make up between 20% and 30% of the minerals used in a mineral-hydrogel mix. This addition of minerals enhances not only the mechanical properties, but also the biological ones. On the one hand, the higher the amount of minerals added to the composition, the better the mechanical properties obtained. Additionally, as the proportion of CaCO3 in comparison with CaHPO4 rises, the mechanical properties improve. On the other hand, both cell proliferation and mineralization are improved with the addition of calcium minerals.
Collapse
Affiliation(s)
- Aitor Tejo-Otero
- Bioengineering Research Group, University of Nottingham, University Park Campus, Nottingham, UK
| | - Alastair C Ritchie
- Bioengineering Research Group, University of Nottingham, University Park Campus, Nottingham, UK
| |
Collapse
|
22
|
Alipour M, Firouzi N, Aghazadeh Z, Samiei M, Montazersaheb S, Khoshfetrat AB, Aghazadeh M. The osteogenic differentiation of human dental pulp stem cells in alginate-gelatin/Nano-hydroxyapatite microcapsules. BMC Biotechnol 2021; 21:6. [PMID: 33430842 PMCID: PMC7802203 DOI: 10.1186/s12896-020-00666-3] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Accepted: 12/22/2020] [Indexed: 02/06/2023] Open
Abstract
Background Microcapsule is considered as a promising 3D microenvironment for Bone Tissue Engineering (BTE) applications. Microencapsulation of cells in an appropriate scaffold not only protected the cells against excess stress but also promoted cell proliferation and differentiation. Through the current study, we aimed to microcapsulate the human Dental Pulp Stem Cells (hDPSCs) and evaluated the proliferation and osteogenic differentiation of those cells by using MTT assay, qRT-PCR, Alkaline phosphatase, and Alizarine Red S. Results The SEM results revealed that Alg/Gel microcapsules containing nHA showed a rough and more compact surface morphology in comparison with the Alg/Gel microcapsules. Moreover, the microencapsulation by Alg/Gel/nHA could improve cell proliferation and induce osteogenic differentiation. The cells cultured in the Alg/Gel and Alg/Gel/nHA microcapsules showed 1.4-fold and 1.7-fold activity of BMP-2 gene expression more in comparison with the control group after 21 days. The mentioned amounts for the BMP-2 gene were 2.5-fold and 4-fold more expression for the Alg/Gel and Alg/Gel/nHA microcapsules after 28 days. The nHA, addition to hDPSCs-laden Alg/Gel microcapsule, could up-regulate the bone-related gene expressions of osteocalcin, osteonectin, and RUNX-2 during the 21 and 28 days through the culturing period, too. Calcium deposition and ALP activities of the cells were observed in accordance with the proliferation results as well as the gene expression analysis. Conclusion The present study demonstrated that microencapsulation of the hDPSCs inside the Alg/Gel/nHA hydrogel could be a potential approach for regenerative dentistry in the near future. Graphical abstract ![]()
Collapse
Affiliation(s)
- Mahdieh Alipour
- Dental and Periodontal Research Center, Faculty of Dentistry, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nima Firouzi
- Stem Cell and Tissue Engineering Research Laboratory, Sahand University of Technology, Tabriz, Iran
| | - Zahra Aghazadeh
- Stem Cell Research Center and Department of Oral Medicine, Faculty of Dentistry, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Samiei
- Department of Endodontics, Faculty of Dentistry, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Soheila Montazersaheb
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Baradar Khoshfetrat
- Stem Cell and Tissue Engineering Research Laboratory, Sahand University of Technology, Tabriz, Iran.
| | - Marziyeh Aghazadeh
- Stem Cell Research Center and Department of Oral Medicine, Faculty of Dentistry, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
23
|
Rafieemehr H, Maleki Behzad M, Azandeh S, Farshchi N, Ghasemi Dehcheshmeh M, Saki N. Chemo/radiotherapy-Induced Bone Marrow Niche Alterations. Cancer Invest 2020; 39:180-194. [PMID: 33225760 DOI: 10.1080/07357907.2020.1855353] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Bone marrow (BM) niche is a specific microenvironment for hematopoietic stem cells (HSCs) as well as non-hematopoietic cells. Evidence shows that chemo/radiotherapy can lead to the disruption of different properties of HSCs such as proliferation, differentiation, localization, self-renewa, and steady-state of cell populations. Investigations have shown that the deregulation of balance within the marrow cavity due to chemo/radiotherapy could lead to bone loss, abnormal hematopoiesis, and enhanced differentiation potential of mesenchymal stem cells towards the adipogenic lineage. Therefore, understanding the underlying mechanisms of chemo/radiotherapy induced BM niche changes may lead to the application of appropriate therapeutic agents to prevent BM niche defects. Highlights Chemo/radiotherapy disrupts the steady-state of bone marrow niche cells and result in deregulation of normal balance of stromal cell populations. Chemo/radiotherapy agents play a significant role in reducing of bone formation as well as fat accumulation in the bone marrow niche. Targeting molecular pathways may lead to recovery of bone marrow niches after chemo/radiotherapy.
Collapse
Affiliation(s)
- Hassan Rafieemehr
- Department of Medical Laboratory Sciences, School of Paramedicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Masumeh Maleki Behzad
- Thalassemia & Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.,Blood Transfusion Research Center, High Institute for Research and Education in Transfusion, Hamadan, Iran
| | - Saeed Azandeh
- Cellular and Molecular Research Center (CMRC), Department of Anatomical Sciences, Faculty of Medicin, Ahvaz Jundishapur University of Medical Sciences (AJUMS), Ahvaz, Iran
| | - Niloofar Farshchi
- Thalassemia & Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | | | - Najmaldin Saki
- Thalassemia & Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
24
|
O’Doherty M, Mulholland EJ, Chambers P, Pentlavalli S, Ziminska M, Chalanqui MJ, Pauly HM, Sathy BN, Donahue TH, Kelly DJ, Dunne N, McCarthy HO. Improving the Intercellular Uptake and Osteogenic Potency of Calcium Phosphate via Nanocomplexation with the RALA Peptide. NANOMATERIALS (BASEL, SWITZERLAND) 2020; 10:E2442. [PMID: 33297306 PMCID: PMC7762210 DOI: 10.3390/nano10122442] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 11/26/2020] [Accepted: 11/27/2020] [Indexed: 11/17/2022]
Abstract
Calcium phosphate-base materials (e.g., alpha tri-calcium phosphate (α-TCP)) have been shown to promote osteogenic differentiation of stem/progenitor cells, enhance osteoblast osteogenic activity and mediate in vivo bone tissue formation. However, variable particle size and hydrophilicity of the calcium phosphate result in an extremely low bioavailability. Therefore, an effective delivery system is required that can encapsulate the calcium phosphate, improve cellular entry and, consequently, elicit a potent osteogenic response in osteoblasts. In this study, collagenous matrix deposition and extracellular matrix mineralization of osteoblast lineage cells were assessed to investigate osteogenesis following intracellular delivery of α-TCP nanoparticles. The nanoparticles were formed via condensation with a novel, cationic 30 mer amphipathic peptide (RALA). Nanoparticles prepared at a mass ratio of 5:1 demonstrated an average particle size of 43 nm with a zeta potential of +26 mV. The average particle size and zeta potential remained stable for up to 28 days at room temperature and across a range of temperatures (4-37 °C). Cell viability decreased 24 h post-transfection following RALA/α-TCP nanoparticle treatment; however, recovery ensued by Day 7. Immunocytochemistry staining for Type I collagen up to Day 21 post-transfection with RALA/α-TCP nanoparticles (NPs) in MG-63 cells exhibited a significant enhancement in collagen expression and deposition compared to an untreated control. Furthermore, in porcine mesenchymal stem cells (pMSCs), there was enhanced mineralization compared to α-TCP alone. Taken together these data demonstrate that internalization of RALA/α-TCP NPs elicits a potent osteogenic response in both MG-63 and pMSCs.
Collapse
Affiliation(s)
- Michelle O’Doherty
- School of Pharmacy, Queen’s University Belfast, Belfast BT9 7BL, UK; (M.O.); (E.J.M.); (P.C.); (S.P.); (M.Z.); (M.J.C.)
| | - Eoghan J. Mulholland
- School of Pharmacy, Queen’s University Belfast, Belfast BT9 7BL, UK; (M.O.); (E.J.M.); (P.C.); (S.P.); (M.Z.); (M.J.C.)
| | - Philip Chambers
- School of Pharmacy, Queen’s University Belfast, Belfast BT9 7BL, UK; (M.O.); (E.J.M.); (P.C.); (S.P.); (M.Z.); (M.J.C.)
| | - Sreekanth Pentlavalli
- School of Pharmacy, Queen’s University Belfast, Belfast BT9 7BL, UK; (M.O.); (E.J.M.); (P.C.); (S.P.); (M.Z.); (M.J.C.)
| | - Monika Ziminska
- School of Pharmacy, Queen’s University Belfast, Belfast BT9 7BL, UK; (M.O.); (E.J.M.); (P.C.); (S.P.); (M.Z.); (M.J.C.)
| | - Marine J. Chalanqui
- School of Pharmacy, Queen’s University Belfast, Belfast BT9 7BL, UK; (M.O.); (E.J.M.); (P.C.); (S.P.); (M.Z.); (M.J.C.)
| | - Hannah M. Pauly
- Department of Biomedical Engineering, University Colorado State University, 1374 Campus Delivery, Fort Collins, CO 80523, USA; (H.M.P.); (T.H.D.)
| | - Binulal N. Sathy
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland; (B.N.S.); (D.J.K.)
| | - Tammy H. Donahue
- Department of Biomedical Engineering, University Colorado State University, 1374 Campus Delivery, Fort Collins, CO 80523, USA; (H.M.P.); (T.H.D.)
- School of Biomedical Engineering, University of Massachusetts Amherst, 130 Natural Resources Road, Amherst, MA 01003, USA
| | - Daniel J. Kelly
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland; (B.N.S.); (D.J.K.)
- Department of Mechanical and Manufacturing Engineering, School of Engineering, Trinity College Dublin, Dublin 2, Ireland
- Department of Anatomy, Royal College of Surgeons in Ireland, Dublin 2, Ireland
- Advanced Materials and Bioengineering Research Centre (AMBER), Royal College of Surgeons in Ireland and Trinity College Dublin, Dublin 2, Ireland
| | - Nicholas Dunne
- School of Pharmacy, Queen’s University Belfast, Belfast BT9 7BL, UK; (M.O.); (E.J.M.); (P.C.); (S.P.); (M.Z.); (M.J.C.)
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland; (B.N.S.); (D.J.K.)
- Department of Mechanical and Manufacturing Engineering, School of Engineering, Trinity College Dublin, Dublin 2, Ireland
- Advanced Materials and Bioengineering Research Centre (AMBER), Royal College of Surgeons in Ireland and Trinity College Dublin, Dublin 2, Ireland
- School of Mechanical and Manufacturing Engineering, Dublin City University, Dublin 9, Ireland
- Centre for Medical Engineering Research, School of Mechanical and Manufacturing Engineering, Dublin City University, Dublin 9, Ireland
- Advanced Manufacturing Research Centre (I-Form), School of Mechanical and Manufacturing Engineering, Dublin City University, Glasnevin, Dublin 9, Ireland
- Advanced Processing Technology Research Centre, Dublin City University, Dublin 9, Ireland
| | - Helen O. McCarthy
- School of Pharmacy, Queen’s University Belfast, Belfast BT9 7BL, UK; (M.O.); (E.J.M.); (P.C.); (S.P.); (M.Z.); (M.J.C.)
- School of Chemical Sciences, Dublin City University, Dublin 9, Ireland
| |
Collapse
|
25
|
Shafiee A, Kehtari M, Zarei Z, Soleimani M, Varshochian R, Ahmadi A, Atyabi F, Dinarvand R. An in situ hydrogel-forming scaffold loaded by PLGA microspheres containing carbon nanotube as a suitable niche for neural differentiation. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 120:111739. [PMID: 33545882 DOI: 10.1016/j.msec.2020.111739] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 11/10/2020] [Accepted: 11/16/2020] [Indexed: 02/06/2023]
Abstract
The cell-extracellular matrix (ECM) interactions are known to have a strong impact on cell behaviors in neural tissues. Due to complex physiology system and limited regenerative capacity of nervous system, neural tissue engineering has attracted attention as a promising strategy. In this study, we designed a hydrogel loaded by poly (lactic-co-glycolic acid) (PLGA) microspheres containing carbon nanotubes (CNT) and the biochemical differentiation factors, as a scaffold, in order to replicate the neural niche for stem cell growth (and/or differentiation). Different formulations from Hyaluronic acid (H), Poloxamer (P), Ethoxy-silane-capped poloxamer (PE), and cross-linked Alginate (Alg) were utilized as an in situ gel structure matrix to mirror the mechanical properties of the ECM of CNS. Subsequently, conductivity, surface morphology, size of microspheres, and CNT dispersion in microsphere were measured using two probes electrical conductometer, scanning electron microscopy (SEM), dynamic light scattering (DLS), and Raman spectroscopy, respectively. According to SEM and fluorescent microscopy images, CNTs increased the porosity of polymeric structure, which, in turn, facilitated the adhesion of stem cells on the surface of microspheres compared with control. Microstructure and rheological behaviors of different gel compositions were investigated using SEM and parallel-plate oscillatory rheometer, respectively. The MTT assay showed the toxicity profile of hydrogels was appropriate for cell transplantation. The confocal images illustrated the 3D platform of P15%H10% and P20%H5% gel formulations containing the PLGA-CNT microspheres, which allows the proliferation of neural stem cells (NSCs) derived from MSC. The results of real-time PCR and immunocytochemistry showed neuronal differentiation capacity of cultured NSCs derived from MSC in the alginate gel that contained PLGA-CNT microspheres as well as other control groups. The dispersion of the CNT-PLGA microspheres, covered by NSCs, into alginate gel in the presence of induction factors was found to notably enhance the expression of Sox2-SYP and β-Tubulin III neuronal markers.
Collapse
Affiliation(s)
- Akram Shafiee
- Department of Pharmaceutical Nanotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran 1417614411, Iran
| | - Mousa Kehtari
- Department of Stem Cell Biology, Stem Cell Technology Research Center, Tehran, Iran
| | - Zeinab Zarei
- Department of Tissue Engineering and Applied Cell Sciences, Faculty of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Masoud Soleimani
- Department of Hematology and Blood Banking, Faculty of Medicine, Tarbiat Modaress University, Tehran, Iran
| | - Reyhaneh Varshochian
- Nanotechnology Research Centre, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Amirhossein Ahmadi
- Pharmaceutical Sciences Research Center, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Fatemeh Atyabi
- Department of Pharmaceutical Nanotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran 1417614411, Iran; Nanotechnology Research Centre, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.
| | - Rassoul Dinarvand
- Department of Pharmaceutical Nanotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran 1417614411, Iran; Nanotechnology Research Centre, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
26
|
Heo DN, Ayan B, Dey M, Banerjee D, Wee H, Lewis GS, Ozbolat IT. Aspiration-assisted bioprinting of co-cultured osteogenic spheroids for bone tissue engineering. Biofabrication 2020; 13. [PMID: 33059343 DOI: 10.1088/1758-5090/abc1bf] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Accepted: 10/15/2020] [Indexed: 12/18/2022]
Abstract
Conventional top-down approaches in tissue engineering involving cell seeding on scaffolds have been widely used in bone engineering applications. However, scaffold-based bone tissue constructs have had limited clinical translation due to constrains in supporting scaffolds, minimal flexibility in tuning scaffold degradation, and low achievable cell seeding density as compared with native bone tissue. Here, we demonstrate a pragmatic and scalable bottom-up method, inspired from embryonic developmental biology, to build three-dimensional (3D) scaffold-free constructs using spheroids as building blocks. Human umbilical vein endothelial cells (HUVECs) were introduced to human mesenchymal stem cells (hMSCs) (hMSC/HUVEC) and spheroids were fabricated by an aggregate culture system. Bone tissue was generated by induction of osteogenic differentiation in hMSC/HUVEC spheroids for 10 days, with enhanced osteogenic differentiation and cell viability in the core of the spheroids compared to hMSC-only spheroids. Aspiration-assisted bioprinting (AAB) is a new bioprinting technique which allows precise positioning of spheroids (11% with respect to the spheroid diameter) by employing aspiration to lift individual spheroids and bioprint them onto a hydrogel. AAB facilitated bioprinting of scaffold-free bone tissue constructs using the pre-differentiated hMSC/HUVEC spheroids. These constructs demonstrated negligible changes in their shape for two days after bioprinting owing to the reduced proliferative potential of differentiated stem cells. Bioprinted bone tissues showed interconnectivity with actin-filament formation and high expression of osteogenic and endothelial-specific gene factors. This study thus presents a viable approach for 3D bioprinting of complex-shaped geometries using spheroids as building blocks, which can be used for various applications including but not limited to, tissue engineering, organ-on-a-chip and microfluidic devices, drug screening and, disease modeling.
Collapse
Affiliation(s)
| | - Bugra Ayan
- Penn State, University Park, Pennsylvania, UNITED STATES
| | - Madhuri Dey
- Penn State, University Park, Pennsylvania, UNITED STATES
| | | | - Hwabok Wee
- Penn State, Hershey, Pennsylvania, UNITED STATES
| | | | | |
Collapse
|
27
|
Labedz-Maslowska A, Bryniarska N, Kubiak A, Kaczmarzyk T, Sekula-Stryjewska M, Noga S, Boruczkowski D, Madeja Z, Zuba-Surma E. Multilineage Differentiation Potential of Human Dental Pulp Stem Cells-Impact of 3D and Hypoxic Environment on Osteogenesis In Vitro. Int J Mol Sci 2020; 21:ijms21176172. [PMID: 32859105 PMCID: PMC7504399 DOI: 10.3390/ijms21176172] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 08/20/2020] [Accepted: 08/25/2020] [Indexed: 02/06/2023] Open
Abstract
Human dental pulp harbours unique stem cell population exhibiting mesenchymal stem/stromal cell (MSC) characteristics. This study aimed to analyse the differentiation potential and other essential functional and morphological features of dental pulp stem cells (DPSCs) in comparison with Wharton’s jelly-derived MSCs from the umbilical cord (UC-MSCs), and to evaluate the osteogenic differentiation of DPSCs in 3D culture with a hypoxic microenvironment resembling the stem cell niche. Human DPSCs as well as UC-MSCs were isolated from primary human tissues and were subjected to a series of experiments. We established a multiantigenic profile of DPSCs with CD45−/CD14−/CD34−/CD29+/CD44+/CD73+/CD90+/CD105+/Stro-1+/HLA-DR− (using flow cytometry) and confirmed their tri-lineage osteogenic, chondrogenic, and adipogenic differentiation potential (using qRT-PCR and histochemical staining) in comparison with the UC-MSCs. The results also demonstrated the potency of DPSCs to differentiate into osteoblasts in vitro. Moreover, we showed that the DPSCs exhibit limited cardiomyogenic and endothelial differentiation potential. Decreased proliferation and metabolic activity as well as increased osteogenic differentiation of DPSCs in vitro, attributed to 3D cell encapsulation and low oxygen concentration, were also observed. DPSCs exhibiting elevated osteogenic potential may serve as potential candidates for a cell-based product for advanced therapy, particularly for bone repair. Novel tissue engineering approaches combining DPSCs, 3D biomaterial scaffolds, and other stimulating chemical factors may represent innovative strategies for pro-regenerative therapies.
Collapse
Affiliation(s)
- Anna Labedz-Maslowska
- Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 30-387 Krakow, Poland; (A.L.-M.); (N.B.); (A.K.); (S.N.); (Z.M.)
| | - Natalia Bryniarska
- Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 30-387 Krakow, Poland; (A.L.-M.); (N.B.); (A.K.); (S.N.); (Z.M.)
- Department of Experimental Neuroendocrinology, Maj Institute of Pharmacology, Polish Academy of Sciences, 31-343 Krakow, Poland
| | - Andrzej Kubiak
- Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 30-387 Krakow, Poland; (A.L.-M.); (N.B.); (A.K.); (S.N.); (Z.M.)
- Institute of Nuclear Physics, Polish Academy of Sciences, 31-342 Krakow, Poland
| | - Tomasz Kaczmarzyk
- Department of Oral Surgery, Faculty of Medicine, Jagiellonian University Medical College, 31-155 Krakow, Poland;
| | - Malgorzata Sekula-Stryjewska
- Laboratory of Stem Cell Biotechnology, Malopolska Centre of Biotechnology, Jagiellonian University, 30-387 Krakow, Poland;
| | - Sylwia Noga
- Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 30-387 Krakow, Poland; (A.L.-M.); (N.B.); (A.K.); (S.N.); (Z.M.)
- Laboratory of Stem Cell Biotechnology, Malopolska Centre of Biotechnology, Jagiellonian University, 30-387 Krakow, Poland;
| | | | - Zbigniew Madeja
- Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 30-387 Krakow, Poland; (A.L.-M.); (N.B.); (A.K.); (S.N.); (Z.M.)
| | - Ewa Zuba-Surma
- Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 30-387 Krakow, Poland; (A.L.-M.); (N.B.); (A.K.); (S.N.); (Z.M.)
- Correspondence: ; Tel.: +48-12-664-61-80
| |
Collapse
|
28
|
Zhou H, Boys AJ, Harrod JB, Bonassar LJ, Estroff LA. Mineral Distribution Spatially Patterns Bone Marrow Stromal Cell Behavior on Monolithic Bone Scaffolds. Acta Biomater 2020; 112:274-285. [PMID: 32479819 PMCID: PMC7372954 DOI: 10.1016/j.actbio.2020.05.032] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 04/30/2020] [Accepted: 05/25/2020] [Indexed: 11/25/2022]
Abstract
Interfaces between soft tissue and bone are characterized by transitional gradients in composition and structure that mediate substantial changes in mechanical properties. For interfacial tissue engineering, scaffolds with mineral gradients have shown promise in controlling osteogenic behavior of seeded bone marrow stromal cells (bMSCs). Previously, we have demonstrated a 'top-down' method for creating monolithic bone-derived scaffolds with patterned mineral distributions similar to native tissue. In the present work, we evaluated the ability of these scaffolds to pattern osteogenic behavior in bMSCs in basic, osteogenic, and chondrogenic biochemical environments. Immunohistochemical (IHC) and histological stains were used to characterize cellular behavior as a function of local mineral content. Alkaline phosphatase, an early marker of osteogenesis, and osteocalcin, a late marker of osteogenesis, were positively correlated with mineral content in basic, osteogenic, and chondrogenic media. The difference in bMSC behavior between the mineralized and demineralized regions was most pronounced in an basic biochemical environment. In the mineralized regions of the scaffold, osteogenic markers were clearly present as early as 4 days in culture. In osteogenic media, osteogenic behavior was observed across the entire scaffold, whereas in chondrogenic media, there was an overall reduction in osteogenic biomarkers. Overall, these results indicate local mineral content of the scaffold plays a key role in spatially patterning bMSC behavior. Our results can be utilized for the development of interfacial tissue engineered scaffolds and understanding the role of local environment in determining bMSC behavior. STATEMENT OF SIGNIFICANCE: Soft tissue-to-bone interfaces, such as tendon-bone, ligament-bone, and cartilage-bone, are ubiquitous in mammalian musculoskeletal systems. These interfacial tissues have distinct, hierarchically-structured gradients of cellular, biochemical, and materials components. Given the complexity of the biological structures, interfacial tissues present unique challenges for tissue engineering. Here, we demonstrate that material-derived cues can spatially pattern osteogenic behavior in bone marrow stromal cells (bMSCs). Specifically, we observed that when the bMSCs are cultured on bone-derived scaffolds with mineral gradients, cells in contact with higher mineral content display osteogenic behavior at earlier times than those on the unmineralized substrate. The ability to pattern the cellular complexity found in native interfaces while maintaining biologically relevant structures is a key step towards creating engineered tissue interfaces.
Collapse
Affiliation(s)
- Hao Zhou
- Department of Materials Science and Engineering, Cornell University, Ithaca, New York 14853, United States
| | - Alexander J Boys
- Department of Materials Science and Engineering, Cornell University, Ithaca, New York 14853, United States
| | - Jordan B Harrod
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York 14853, United States
| | - Lawrence J Bonassar
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York 14853, United States; Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, New York 14853, United States.
| | - Lara A Estroff
- Department of Materials Science and Engineering, Cornell University, Ithaca, New York 14853, United States; Kavli Institute for Nanoscale Science at Cornell, Cornell University, Ithaca, New York 14853, United States.
| |
Collapse
|
29
|
MiR-34a suppresses osteoblast differentiation through glycolysis inhibition by targeting lactate dehydrogenase-A (LDHA). In Vitro Cell Dev Biol Anim 2020; 56:480-487. [PMID: 32719987 DOI: 10.1007/s11626-020-00467-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Accepted: 05/07/2020] [Indexed: 01/24/2023]
Abstract
Bone formation (osteogenesis) is mediated through recruitment of bone marrow mesenchymal stem cells (MSCs) with capacity to differentiate into osteoblasts, a process which is regulated by transcriptional and post-transcriptional mechanisms. Multiple studies have suggested that miRNAs might have important roles in osteoblast differentiation. Expressions of miR-34a were detected by qRT-PCR. Cellular glucose metabolism was assessed by measurements of glucose uptake and lactate production. mRNA expressions of glycolysis enzymes were detected by qRT-PCR. Osteogenic differentiation of human MSCs (hMSCs) was analyzed by alkaline phosphatase (ALP) activity and Alizarin red staining. Here, we report that microRNA-34a is upregulated during the osteoblast differentiation from hMSCs. miR-34a overexpressing inhibited late osteoblast differentiation of hMSCs in vitro. The ALP activity and Alizarin red staining were significantly decreased by miR-34a in hMSCs. Target prediction analysis reveals that the lactate dehydrogenase-A (LDHA) is a potential target of miR-34a. We hypothesized that miR-34a inhibits osteoblast differentiation through targeting the LDHA-mediated cellular glycolysis. Results from Western blotting and luciferase assay validated that miR-34a could directly target 3'UTR of LDHA mRNA. In addition, we demonstrated that overexpression of miR-34a inhibits cellular anaerobic glycolysis through targeting LHDA. The protein and mRNA expressions of glycolysis enzymes, Hexokinase 2 (HK2), glucose transporter 1 (GLUT1), and LDHA were significantly downregulated by miR-34a overexpression in hMSCs. Furthermore, we showed that LDHA restoration in miR-34a overexpressing hMSCs successfully rescued the osteoblast differentiation of hMSCs. This study demonstrated the roles of miR-34a in regulating osteoblast differentiation, suggesting that miR-34a inhibition could be a new therapeutic strategy for improving bone formation.
Collapse
|
30
|
Filippi M, Born G, Chaaban M, Scherberich A. Natural Polymeric Scaffolds in Bone Regeneration. Front Bioeng Biotechnol 2020; 8:474. [PMID: 32509754 PMCID: PMC7253672 DOI: 10.3389/fbioe.2020.00474] [Citation(s) in RCA: 161] [Impact Index Per Article: 32.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Accepted: 04/23/2020] [Indexed: 12/13/2022] Open
Abstract
Despite considerable advances in microsurgical techniques over the past decades, bone tissue remains a challenging arena to obtain a satisfying functional and structural restoration after damage. Through the production of substituting materials mimicking the physical and biological properties of the healthy tissue, tissue engineering strategies address an urgent clinical need for therapeutic alternatives to bone autografts. By virtue of their structural versatility, polymers have a predominant role in generating the biodegradable matrices that hold the cells in situ to sustain the growth of new tissue until integration into the transplantation area (i.e., scaffolds). As compared to synthetic ones, polymers of natural origin generally present superior biocompatibility and bioactivity. Their assembly and further engineering give rise to a wide plethora of advanced supporting materials, accounting for systems based on hydrogels or scaffolds with either fibrous or porous architecture. The present review offers an overview of the various types of natural polymers currently adopted in bone tissue engineering, describing their manufacturing techniques and procedures of functionalization with active biomolecules, and listing the advantages and disadvantages in their respective use in order to critically compare their actual applicability potential. Their combination to other classes of materials (such as micro and nanomaterials) and other innovative strategies to reproduce physiological bone microenvironments in a more faithful way are also illustrated. The regeneration outcomes achieved in vitro and in vivo when the scaffolds are enriched with different cell types, as well as the preliminary clinical applications are presented, before the prospects in this research field are finally discussed. The collection of studies herein considered confirms that advances in natural polymer research will be determinant in designing translatable materials for efficient tissue regeneration with forthcoming impact expected in the treatment of bone defects.
Collapse
Affiliation(s)
- Miriam Filippi
- Department of Biomedical Engineering, University of Basel, Basel, Switzerland.,Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Gordian Born
- Department of Biomedical Engineering, University of Basel, Basel, Switzerland
| | - Mansoor Chaaban
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Arnaud Scherberich
- Department of Biomedical Engineering, University of Basel, Basel, Switzerland.,Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| |
Collapse
|
31
|
Silanization improves biocompatibility of graphene oxide. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 110:110647. [DOI: 10.1016/j.msec.2020.110647] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 06/19/2019] [Accepted: 01/03/2020] [Indexed: 11/21/2022]
|
32
|
Yu L, Rowe DW, Perera IP, Zhang J, Suib SL, Xin X, Wei M. Intrafibrillar Mineralized Collagen-Hydroxyapatite-Based Scaffolds for Bone Regeneration. ACS APPLIED MATERIALS & INTERFACES 2020; 12:18235-18249. [PMID: 32212615 DOI: 10.1021/acsami.0c00275] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
As one of the major challenges in the field of tissue engineering, large skeletal defects have attracted wide attention from researchers. Collagen (Col) and hydroxyapatite (HA), the most abundant protein and the main component in natural bone, respectively, are usually used as a biomimetic composite material in tissue engineering due to their excellent biocompatibility and biodegradability. In this study, novel intrafibrillar mineralized Col-HA-based scaffolds, constructed in either cellular or lamellar microstructures, were established through a biomimetic method to enhance the new bone-regenerating capability of tissue engineering scaffolds. Moreover, iron (Fe) and manganese (Mn), two of the essential trace elements in the body, were successfully incorporated into the lamellar scaffold to further improve the osteoinductivity of these biomaterials. It was found that the lamellar scaffolds demonstrated better osteogenic abilities compared to both in-house and commercial Col-HA-based cellular scaffolds in vitro and in vivo. Meanwhile, Fe/Mn incorporation further amplified the osteogenic promotion of the lamellar scaffolds. More importantly, a synergistic effect was observed in the Fe and Mn dual-element-incorporated lamellar scaffolds for both in vitro osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs) and in vivo bone regeneration loaded with fresh bone marrow cells. This study provides a simple but practical strategy for the creation of functional scaffolds for bone regeneration.
Collapse
Affiliation(s)
- Le Yu
- Department of Chemical and Biomolecular Engineering, Ohio University, Athens, Ohio 45701, United States
| | - David W Rowe
- Center for Regenerative Medicine and Skeletal Development, School of Dental Medicine, University of Connecticut Health Center, Farmington, Connecticut 06032, United States
| | | | | | | | - Xiaonan Xin
- Center for Regenerative Medicine and Skeletal Development, School of Dental Medicine, University of Connecticut Health Center, Farmington, Connecticut 06032, United States
| | - Mei Wei
- Department of Mechanical Engineering, Ohio University, Athens, Ohio 45701, United States
| |
Collapse
|
33
|
Schwieger J, Hamm A, Gepp MM, Schulz A, Hoffmann A, Lenarz T, Scheper V. Alginate-encapsulated brain-derived neurotrophic factor-overexpressing mesenchymal stem cells are a promising drug delivery system for protection of auditory neurons. J Tissue Eng 2020; 11:2041731420911313. [PMID: 32341778 PMCID: PMC7168777 DOI: 10.1177/2041731420911313] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 02/08/2020] [Indexed: 12/23/2022] Open
Abstract
The cochlear implant outcome is possibly improved by brain-derived neurotrophic factor treatment protecting spiral ganglion neurons. Implantation of genetically modified mesenchymal stem cells may enable the required long-term brain-derived neurotrophic factor administration. Encapsulation of mesenchymal stem cells in ultra-high viscous alginate may protect the mesenchymal stem cells from the recipient’s immune system and prevent their uncontrolled migration. Alginate stability and survival of mesenchymal stem cells in alginate were evaluated. Brain-derived neurotrophic factor production was measured and its protective effect was analyzed in dissociated rat spiral ganglion neuron co-culture. Since the cochlear implant is an active electrode, alginate–mesenchymal stem cell samples were electrically stimulated and alginate stability and mesenchymal stem cell survival were investigated. Stability of ultra-high viscous-alginate and alginate–mesenchymal stem cells was proven. Brain-derived neurotrophic factor production was detectable and spiral ganglion neuron survival, bipolar morphology, and neurite outgrowth were increased. Moderate electrical stimulation did not affect the mesenchymal stem cell survival and their viability was good within the investigated time frame. Local drug delivery by ultra-high viscous-alginate-encapsulated brain-derived neurotrophic factor–overexpressing mesenchymal stem cells is a promising strategy to improve the cochlear implant outcome.
Collapse
Affiliation(s)
- Jana Schwieger
- Department of Otolaryngology, Hannover Medical School, Hannover, Germany.,NIFE-Lower Saxony Centre for Biomedical Engineering, Implant Research and Development, Hannover, Germany
| | - Anika Hamm
- NIFE-Lower Saxony Centre for Biomedical Engineering, Implant Research and Development, Hannover, Germany.,Department of Orthopaedic Surgery, Hannover Medical School, Hannover, Germany
| | - Michael M Gepp
- Fraunhofer Institute for Biomedical Engineering IBMT, Sulzbach, Germany.,Fraunhofer Project Center for Stem Cell Process Engineering, Würzburg, Germany
| | - André Schulz
- Fraunhofer Institute for Biomedical Engineering IBMT, Sulzbach, Germany
| | - Andrea Hoffmann
- NIFE-Lower Saxony Centre for Biomedical Engineering, Implant Research and Development, Hannover, Germany.,Department of Orthopaedic Surgery, Hannover Medical School, Hannover, Germany
| | - Thomas Lenarz
- Department of Otolaryngology, Hannover Medical School, Hannover, Germany.,NIFE-Lower Saxony Centre for Biomedical Engineering, Implant Research and Development, Hannover, Germany.,Cluster of Excellence Hearing4all, German Research Foundation, Hannover, Germany
| | - Verena Scheper
- Department of Otolaryngology, Hannover Medical School, Hannover, Germany.,NIFE-Lower Saxony Centre for Biomedical Engineering, Implant Research and Development, Hannover, Germany.,Cluster of Excellence Hearing4all, German Research Foundation, Hannover, Germany
| |
Collapse
|
34
|
May RD, Frauchiger DA, Albers CE, Hofstetter W, Gantenbein B. Exogenous Stimulation of Human Intervertebral Disc Cells in 3-Dimensional Alginate Bead Culture With BMP2 and L51P: Cytocompatibility and Effects on Cell Phenotype. Neurospine 2020; 17:77-87. [PMID: 32252157 PMCID: PMC7136110 DOI: 10.14245/ns.2040002.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Accepted: 02/18/2020] [Indexed: 12/16/2022] Open
Abstract
Objective Spinal fusion surgery is a common treatment modality for various pathologic conditions of the spine. The bone morphogenetic protein 2 (BMP2) analogue L51P acts as a general inhibitor of BMP antagonists, whereas it shows a weak affinity for BMP type I receptor. It is suggested that L51P applied in bone disorders might prevent side effects of highly concentrated BMP dosage applications in the order of milligrams. The objective of this study was to investigate the effects of L51P and BMP2 on intervertebral disc cells (IVDCs), i.e. on nucleus pulposus cells, on annulus fibrosus cells (AFCs), and on cartilaginous endplate cells (CEPCs), respectively, in 3-dimensional (3D) culture.
Methods Low-passage primary IVDCs were cultured in 3D alginate bead culture and exposed to 100-ng/mL BMP2 and/or L51P for 21 days. Here, we analyzed glycosaminoglycan (GAG) and DNA content and further performed gene expression analysis for major matrix genes.
Results AFCs and cartilaginous CEPCs stimulated with each 100-ng/mL L51P and BMP2, showed a significant upregulation in GAG (AFCs: p = 0.00347 and CEPCs: p = 0.0115) and DNA production (AFCs: p = 0.0182 and CEPCs: p = 0.0179) compared to control.
Conclusion These results allow first insights into the behavior of IVDCs upon L51P stimulation.
Collapse
Affiliation(s)
- Rahel D May
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Daniela A Frauchiger
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Christoph E Albers
- Department of Orthopedic Surgery and Traumatology, Inselspital Bern, University of Bern, Bern, Switzerland
| | - Willy Hofstetter
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Benjamin Gantenbein
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland.,Department of Orthopedic Surgery and Traumatology, Inselspital Bern, University of Bern, Bern, Switzerland
| |
Collapse
|
35
|
Ayan B, Heo DN, Zhang Z, Dey M, Povilianskas A, Drapaca C, Ozbolat IT. Aspiration-assisted bioprinting for precise positioning of biologics. SCIENCE ADVANCES 2020; 6:eaaw5111. [PMID: 32181332 PMCID: PMC7060055 DOI: 10.1126/sciadv.aaw5111] [Citation(s) in RCA: 162] [Impact Index Per Article: 32.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 12/13/2019] [Indexed: 05/13/2023]
Abstract
Three-dimensional (3D) bioprinting is an appealing approach for building tissues; however, bioprinting of mini-tissue blocks (i.e., spheroids) with precise control on their positioning in 3D space has been a major obstacle. Here, we unveil "aspiration-assisted bioprinting (AAB)," which enables picking and bioprinting biologics in 3D through harnessing the power of aspiration forces, and when coupled with microvalve bioprinting, it facilitated different biofabrication schemes including scaffold-based or scaffold-free bioprinting at an unprecedented placement precision, ~11% with respect to the spheroid size. We studied the underlying physical mechanism of AAB to understand interactions between aspirated viscoelastic spheroids and physical governing forces during aspiration and bioprinting. We bioprinted a wide range of biologics with dimensions in an order-of-magnitude range including tissue spheroids (80 to 600 μm), tissue strands (~800 μm), or single cells (electrocytes, ~400 μm), and as applications, we illustrated the patterning of angiogenic sprouting spheroids and self-assembly of osteogenic spheroids.
Collapse
Affiliation(s)
- Bugra Ayan
- Engineering Science and Mechanics Department, Penn State University, University Park, PA 16802, USA
- The Huck Institutes of the Life Sciences, Penn State University, University Park, PA 16802, USA
| | - Dong Nyoung Heo
- Engineering Science and Mechanics Department, Penn State University, University Park, PA 16802, USA
- The Huck Institutes of the Life Sciences, Penn State University, University Park, PA 16802, USA
- Department of Dental Materials, School of Dentistry, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Zhifeng Zhang
- Engineering Science and Mechanics Department, Penn State University, University Park, PA 16802, USA
| | - Madhuri Dey
- The Huck Institutes of the Life Sciences, Penn State University, University Park, PA 16802, USA
- Department of Chemistry, Penn State University, University Park, PA 16802, USA
| | - Adomas Povilianskas
- Engineering Science and Mechanics Department, Penn State University, University Park, PA 16802, USA
| | - Corina Drapaca
- Engineering Science and Mechanics Department, Penn State University, University Park, PA 16802, USA
| | - Ibrahim T. Ozbolat
- Engineering Science and Mechanics Department, Penn State University, University Park, PA 16802, USA
- The Huck Institutes of the Life Sciences, Penn State University, University Park, PA 16802, USA
- Biomedical Engineering Department, Penn State University, University Park, PA 16802, USA
- Materials Research Institute, Penn State University, University Park, PA 16802, USA
| |
Collapse
|
36
|
The fate of mesenchymal stem cells is greatly influenced by the surface chemistry of silica nanoparticles in 3D hydrogel-based culture systems. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 106:110259. [DOI: 10.1016/j.msec.2019.110259] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 09/18/2019] [Accepted: 09/25/2019] [Indexed: 12/17/2022]
|
37
|
Merkel A, Chen Y, George A. Endocytic Trafficking of DMP1 and GRP78 Complex Facilitates Osteogenic Differentiation of Human Periodontal Ligament Stem Cells. Front Physiol 2019; 10:1175. [PMID: 31572220 PMCID: PMC6751249 DOI: 10.3389/fphys.2019.01175] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Accepted: 08/30/2019] [Indexed: 12/22/2022] Open
Abstract
Periodontal ligament contains periodontal ligament stem cells that maintain tissue homeostasis. Targeting hPDLSCs (human periodontal ligament cells) is a promising strategy for repair and regeneration of bone tissue destroyed by periodontal diseases. However, the mechanisms by which PDLSCs differentiate into osteoblasts to form a mineralized matrix is unclear. In this study, we demonstrate for the first time the molecular events that contribute to osteogenic differentiation of PDLSCs. Dentin matrix protein 1 (DMP1) and its receptor, Glucose regulated protein-78 (GRP78), are localized in the progenitor cells of the PDL. Our overall goal is to demonstrate the formation of DMP1-GRP78 complex at the plasma membrane and subsequent protein trafficking and nuclear localization to promote osteogenic differentiation. To study the internalization and routing of the complex, we mimic an in vivo differentiation scenario by stimulating cells with DMP1 and culturing them in the presence of osteogenic differentiation conditions. We first demonstrate the translocation of the ER chaperone protein GRP78 to the plasma membrane during the differentiation process. Total internal reflection microscopy imaging demonstrates the formation and internalization of the receptor- ligand (GRP78-DMP1) complex. Confocal microscopy results show the internalization of the GRP78-DMP1 complex specifically through the caveolin pathway and trafficked through the cell with various endocytic markers such as Rab5 and 7 GTPases to early and late endosomes respectively. DMP1 is ultimately transported to the nucleus where it functions to promote osteogenic differentiation as demonstrated by quantitative Real-Time PCR. This observation is the first report that suggests DMP1 and GRP78 can interact at the plasma membrane, then packaged in vesicles and ultimately DMP1 is routed to the nucleus where it aids in osteogenic differentiation of PDLSCs. Characterizing the osteogenic potential of PDLSCs would favor the development of therapeutic strategies for reconstruction of mineralized tissues destroyed by periodontal diseases.
Collapse
Affiliation(s)
| | | | - Anne George
- Department of Oral Biology, The University of Illinois at Chicago, Chicago, IL, United States
| |
Collapse
|
38
|
Pan Y, Li Z, Wang Y, Yan M, Wu J, Beharee RG, Yu J. Sodium fluoride regulates the osteo/odontogenic differentiation of stem cells from apical papilla by modulating autophagy. J Cell Physiol 2019; 234:16114-16124. [PMID: 30767218 DOI: 10.1002/jcp.28269] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 01/18/2019] [Accepted: 01/22/2019] [Indexed: 01/24/2023]
Abstract
Fluoride (sodium fluoride) is thought to be essential in the development of tooth, and research shows that fluoride can modulate the differentiation of dental stem cells. However, the effects of fluoride on the committed differentiation of stem cells from apical papilla (SCAPs) and the underlying mechanisms remain unclear. Here, SCAPs were isolated from healthy extracted human third molars with immature roots and then were cultured with NaF conditioned media. Cell Counting Kit-8, EdU staining, and flow cytometry were performed to detected the proliferation activity. Alkaline phosphatase (ALP) activity, Alizarin Red staining, Western blot assay, and real-time reverse-transcription polymerase chain reaction were applied to assess the osteo/odontogenic differentiation NaF-treated SCAPs. Western blot assay and transmission electron microscope were used to evaluate the autophagy involved in the differentiation of SCAPs. ALP activity, ALP protein, and messenger RNA (mRNA) expression showed that 0.5 mM was the optimal concentration for the induction of SCAPs by NaF. 0.5 mM NaF-treated SCAPs induced more mineralized nodules as compared with untreated cells. Moreover, the osteo/odontogenic markers (RUNX2, OSX, DSP, and OCN) in mRNA levels were upregulated while the protein levels of these markers increased considerably in 0.5 mM NaF-treated SCAPs. Furthermore, the autophagy-related proteins (LC3, ATG5, and Beclin1) increased in NaF-treated SCAPs, and the osteo/odontogenic makers significantly decreased while silencing ATG5 to block autophagy. In all, sodium fluoride can regulate the osteo/odontogenic differentiation of SCAPs by modulating autophagy.
Collapse
Affiliation(s)
- Yin Pan
- Key Laboratory of Oral Diseases of Jiangsu Province, Institute of Stomatology, Nanjing Medical University, Nanjing, Jiangsu, China.,Department of Endodontics, School of Stomatology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Zehan Li
- Key Laboratory of Oral Diseases of Jiangsu Province, Institute of Stomatology, Nanjing Medical University, Nanjing, Jiangsu, China.,Department of Endodontics, School of Stomatology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yanqiu Wang
- Key Laboratory of Oral Diseases of Jiangsu Province, Institute of Stomatology, Nanjing Medical University, Nanjing, Jiangsu, China.,Department of Endodontics, School of Stomatology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Ming Yan
- Key Laboratory of Oral Diseases of Jiangsu Province, Institute of Stomatology, Nanjing Medical University, Nanjing, Jiangsu, China.,Department of Endodontics, School of Stomatology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jintao Wu
- Key Laboratory of Oral Diseases of Jiangsu Province, Institute of Stomatology, Nanjing Medical University, Nanjing, Jiangsu, China.,Department of Endodontics, School of Stomatology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Romila Gobin Beharee
- Key Laboratory of Oral Diseases of Jiangsu Province, Institute of Stomatology, Nanjing Medical University, Nanjing, Jiangsu, China.,Department of Endodontics, School of Stomatology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jinhua Yu
- Key Laboratory of Oral Diseases of Jiangsu Province, Institute of Stomatology, Nanjing Medical University, Nanjing, Jiangsu, China.,Department of Endodontics, School of Stomatology, Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
39
|
Pasternak-Mnich K, Ziemba B, Szwed A, Kopacz K, Synder M, Bryszewska M, Kujawa J. Effect of Photobiomodulation Therapy on the Increase of Viability and Proliferation of Human Mesenchymal Stem Cells. Lasers Surg Med 2019; 51:824-833. [PMID: 31165521 DOI: 10.1002/lsm.23107] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/07/2019] [Indexed: 12/13/2022]
Abstract
BACKGROUND AND OBJECTIVES We have investigated how low intensity laser irradiation emitted by a multiwave-locked system (MLS M1) affects the viability and proliferation of human bone marrow mesenchymal stem cells (MSCs) depending on the parameters of the irradiation. STUDY DESIGN/MATERIALS AND METHODS Cells isolated surgically from the femoral bone during surgery were identified by flow cytometry and cell differentiation assays. For irradiation, two wavelengths (808 and 905 nm) with the following parameters were used: power density 195, 230, and 318 mW/cm 2 , doses of energy 3, 10, and 20 J (energy density 0.93-6.27 J/cm 2 ), and in continuous (CW) or pulsed emission (PE) (frequencies 1,000 and 2,000 Hz). RESULTS There were statistically significant increases of cell viability and proliferation after irradiation at 3 J (CW; 1,000 Hz), 10 J (1,000 Hz), and 20 J (2,000 Hz). CONCLUSIONS Irradiation with the MLS M1 system can be used in vitro to modulate MSCs in preparation for therapeutic applications. This will assist in designing further studies to optimize the radiation parameters and elucidate the molecular mechanisms of action of the radiation. Lasers Surg. Med. © 2019 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Kamila Pasternak-Mnich
- Department of Medical Rehabilitation, Faculty of Health Sciences, Medical University of Lodz, 251 Pomorska St., 92-213, Lodz, Poland
| | - Barbara Ziemba
- Department of Clinical Genetic, Medical University of Lodz, 251 Pomorska St., 92-213, Lodz, Poland
| | - Aleksandra Szwed
- Department of General Biophysics, University of Lodz, 141/143 Pomorska St., 90-236, Lodz, Poland
| | - Karolina Kopacz
- "DynamoLab" Academic Laboratory of Movement and Human Physical Performance, Medical University of Lodz, 251 Pomorska St., 92-213, Lodz, Poland
| | - Marek Synder
- Medical Faculty, Clinic of Orthopedics and Pediatric Orthopedics, Medical University of Lodz, 251 Pomorska St., 92-213, Lodz, Poland
| | - Maria Bryszewska
- Department of General Biophysics, University of Lodz, 141/143 Pomorska St., 90-236, Lodz, Poland
| | - Jolanta Kujawa
- Department of Medical Rehabilitation, Faculty of Health Sciences, Medical University of Lodz, 251 Pomorska St., 92-213, Lodz, Poland
| |
Collapse
|
40
|
Trucillo E, Bisceglia B, Valdrè G, Giordano E, Reverchon E, Maffulli N, Della Porta G. Growth factor sustained delivery from poly-lactic-co-glycolic acid microcarriers and its mass transfer modeling by finite element in a dynamic and static three-dimensional environment bioengineered with stem cells. Biotechnol Bioeng 2019; 116:1777-1794. [PMID: 30905072 DOI: 10.1002/bit.26975] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 02/18/2019] [Accepted: 03/21/2019] [Indexed: 12/15/2022]
Abstract
Poly-lactic-co-glycolic acid (PLGA) microcarriers (0.8 ± 0.2 μm) have been fabricated with a load of 20 μg/gPLGA by an emulsion-based-proprietary technology to sustained deliver human bone morphogenetic protein 2 (hBMP2), a growth factor largely used for osteogenic induction. hBMP2 release profile, measured in vitro, showed a moderate "burst" release of 20% of the load in first 3 days, followed by a sustained release of 3% of the load along the following 21 days. PLGA microbeads loaded with fluorescent marker (8 mg/gPLGA ) and hydroxyapatite (30 mg/gPLGA ) were also fabricated and successfully dispersed within three-dimensional (3D) alginate scaffold (Ca-alginate 2% wt/wt) in a range between 50 and 200 mg/cm3 ; the presence of microcarriers within the scaffold induced a variation of its stiffness between 0.03 and 0.06 MPa; whereas the scaffold surface area was monitored always in the range of 190-200 m2 /g. Uniform microcarriers dispersion was obtained up to 200 mg/cm3 ; higher loading values in the 3D scaffold produced large aggregates. The release data and the surface area were, then, used to simulate by finite element modeling the hBMP2 mass transfer within the 3D hydrogel bioengineered with stem cells, in dynamic and static cultivations. The simulation was developed with COMSOL Multiphysics® giving a good representation of hBMP2 mass balances along microbeads (bulk eroded) and on cell surface (cell binding). hBMP2 degradation rate was also taken into account in the simulations. hBMP2 concentration of 20 ng/cm3 was set as a target because it has been described as the minimum effective value for stem cells stimulation versus the osteogenic phenotype. The sensitivity analysis suggested the best microbeads/cells ratio in the 3D microenvironment, along 21 days of cultivations in both static and dynamic cultivation (perfusion) conditions. The simulated formulation was so assembled experimentally using human mesenchymal stem cells and an improved scaffold stiffness up to 0.09 MPa (n = 3; p ≤ 0.01) was monitored after 21 days of cultivation; moreover a uniform extracellular matrix deposition within the 3D system was detected by Von Kossa staining, especially in dynamic conditions. The results indicated that the described tool can be useful for the design of 3D bioengineered microarchitecture by quantitative understanding.
Collapse
Affiliation(s)
- Emanuele Trucillo
- Department of Industrial Engineering, University of Salerno, Fisciano, SA, Italy
| | - Bruno Bisceglia
- Department of Industrial Engineering, University of Salerno, Fisciano, SA, Italy
| | - Giovanni Valdrè
- Department of Biology, Geology and Environmental Science, University of Bologna, Bologna, BO, Italy
| | - Emanuele Giordano
- Department of Electrical, Electronic and Information Engineering "Guglielmo Marconi" (DEI), University of Bologna, Cesena, FC, Italy
| | - Ernesto Reverchon
- Department of Industrial Engineering, University of Salerno, Fisciano, SA, Italy
| | - Nicola Maffulli
- Translational Medicine Laboratory, Department of Medicine, Surgery and Dentistry, University of Salerno, Baronissi, SA, Italy
| | - Giovanna Della Porta
- Department of Industrial Engineering, University of Salerno, Fisciano, SA, Italy.,Translational Medicine Laboratory, Department of Medicine, Surgery and Dentistry, University of Salerno, Baronissi, SA, Italy
| |
Collapse
|
41
|
Tanimowo Aiyelabegan H, Ebadi M, Ali Kardar G, Lotfibakhshaiesh N, Abedin Dorkoosh F, Ebrahimi_Barough S, Sadroddiny E. k-Casein upregulates osteogenic differentiation on bone marrow mesenchymal stem cells cultured on agarose microcarriers. INT J POLYM MATER PO 2019. [DOI: 10.1080/00914037.2019.1570511] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Affiliation(s)
- Hammed Tanimowo Aiyelabegan
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Malihe Ebadi
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Gholam Ali Kardar
- Immunology, Asthma and Allergy Research Institute, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Nasrin Lotfibakhshaiesh
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Farid Abedin Dorkoosh
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Somayeh Ebrahimi_Barough
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Esmaeil Sadroddiny
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
42
|
Griffanti G, Jiang W, Nazhat SN. Bioinspired mineralization of a functionalized injectable dense collagen hydrogel through silk sericin incorporation. Biomater Sci 2019; 7:1064-1077. [DOI: 10.1039/c8bm01060a] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
The incorporation of silk sericin into injectable dense collagen hydrogels represents a powerful approach to mimic the biomineralization process, together with the osteogenic stimulation of seeded mesenchymal stem cells, in vitro.
Collapse
Affiliation(s)
- Gabriele Griffanti
- Department of Mining and Materials Engineering
- McGill University
- Montréal
- Canada
| | - Wenge Jiang
- Department of Mining and Materials Engineering
- McGill University
- Montréal
- Canada
| | - Showan N. Nazhat
- Department of Mining and Materials Engineering
- McGill University
- Montréal
- Canada
| |
Collapse
|
43
|
Koopaie M. Scaffolds for gingival tissues. HANDBOOK OF TISSUE ENGINEERING SCAFFOLDS: VOLUME ONE 2019:521-543. [DOI: 10.1016/b978-0-08-102563-5.00025-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
44
|
Griffanti G, James-Bhasin M, Donelli I, Freddi G, Nazhat SN. Functionalization of silk fibroin through anionic fibroin derived polypeptides. Biomed Mater 2018; 14:015006. [DOI: 10.1088/1748-605x/aae745] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
45
|
Itel F, Skovhus Thomsen J, Städler B. Matrix Vesicles-Containing Microreactors as Support for Bonelike Osteoblasts to Enhance Biomineralization. ACS APPLIED MATERIALS & INTERFACES 2018; 10:30180-30190. [PMID: 30113809 DOI: 10.1021/acsami.8b10886] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Therapeutic cell mimicry aims to provide a source of cell-like assemblies, which exhibit the core structural or functional properties of their natural counterparts with broad envisioned applications in biomedicine. Bone tissue engineering (BTE) aims at promoting and inciting the natural healing process of, for instance, critically sized bone defects. Microreactors designed to co-assemble with biological bone-forming osteoblasts like SaOS-2 cells to start biomineralization are reported for the first time. The alginate-based microparticles are equipped with active alkaline phosphatase-loaded artificial liposomes or SaOS-2-derived matrix vesicles (MVs). Spheroids assembled from SaOS-2 cells and microreactors not only exhibit higher cell viability, but also show enhanced biomineralization when MVs are present. The active biomineralization stimulation of the microreactors is illustrated by colorimetric calcium quantification and micro-computed tomography. These findings show the promising potential of applying cell mimicry in BTE.
Collapse
Affiliation(s)
- Fabian Itel
- Interdisciplinary Nanoscience Center (iNANO) , Aarhus University , Gustav Wieds Vej 14 , 8000 Aarhus , Denmark
| | - Jesper Skovhus Thomsen
- Department of Biomedicine , Aarhus University , Wilhelm Meyers Allé 3 , 8000 Aarhus , Denmark
| | - Brigitte Städler
- Interdisciplinary Nanoscience Center (iNANO) , Aarhus University , Gustav Wieds Vej 14 , 8000 Aarhus , Denmark
| |
Collapse
|
46
|
Iijima K, Ishikawa S, Sasaki K, Hashizume M, Kawabe M, Otsuka H. Osteogenic Differentiation of Bone Marrow-Derived Mesenchymal Stem Cells in Electrospun Silica Nonwoven Fabrics. ACS OMEGA 2018; 3:10180-10187. [PMID: 31459146 PMCID: PMC6645240 DOI: 10.1021/acsomega.8b01139] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Accepted: 08/20/2018] [Indexed: 05/12/2023]
Abstract
Silica nonwoven fabrics (SNFs) with enough mechanical strength are candidates as implantable scaffolds. Culture of cells therein is expected to affect the proliferation and differentiation of the cells through cell-cell and cell-SNF interactions. In this study, we examined three-dimensional (3D) SNFs as a scaffold of mesenchymal stem cells (MSCs) for bone tissue engineering applications. The interconnected highly porous microstructure of 3D SNFs is expected to allow omnidirectional cell-cell interactions, and the morphological similarity of a silica nanofiber to that of a fibrous extracellular matrix can contribute to the promotion of cell functions. 3D SNFs were prepared by the sol-gel process, and their mechanical properties were characterized by the compression test and rheological analysis. In the compression test, SNFs showed a compressive elastic modulus of over 1 MPa and a compressive strength of about 200 kPa. These values are higher than those of porous polystyrene disks used for in vitro 3D cell culture. In rheological analysis, the elastic modulus and fracture stress were 3.27 ± 0.54 kPa and 25.9 ± 8.3 Pa, respectively. Then, human bone marrow-derived MSCs were cultured on SNFs, and the effects on proliferation and osteogenic differentiation were evaluated. The MSCs seeded on SNF proliferated, and the thickness of the cell layer became over 80 μm after 14 days of culture. The osteogenic differentiation of MSCs on SNFs was induced by the culture in the commercial osteogenic differentiation medium. The alkaline phosphatase activity of MSCs on SNFs increased rapidly and remained high up to 14 days and was much higher than that on two-dimensional tissue culture-treated polystyrene. The high expression of RUNX2 and intense staining by alizarin red s after differentiation supported that SNFs enhanced the osteogenic differentiation of MSCs. Furthermore, permeation analysis of SNFs using fluorescein isothiocyanate-dextran suggested a sufficient permeability of SNFs for oxygen, minerals, nutrients, and secretions, which is important for maintaining the cell viability and vitality. These results suggested that SNFs are promising scaffolds for the regeneration of bone defects using MSCs, originated from highly porous and elastic SNF characters.
Collapse
Affiliation(s)
- Kazutoshi Iijima
- Department
of Industrial Chemistry, Faculty of Engineering, Tokyo University of Science, 12-1 Ichigayafunagawara-machi,
Shinjuku-ku, Tokyo 162-0826, Japan
| | - Shohei Ishikawa
- Graduate School of Science and Department of
Applied Chemistry, Faculty of
Science, Tokyo University of Science, 1-3 Kagurazaka,
Shinjuku-ku, Tokyo 162-8601, Japan
| | - Kohei Sasaki
- Japan
Vilene Company Ltd., 7 kita-Tone, Koga, Ibaraki 306-0213, Japan
| | - Mineo Hashizume
- Department
of Industrial Chemistry, Faculty of Engineering, Tokyo University of Science, 12-1 Ichigayafunagawara-machi,
Shinjuku-ku, Tokyo 162-0826, Japan
| | - Masaaki Kawabe
- Japan
Vilene Company Ltd., 7 kita-Tone, Koga, Ibaraki 306-0213, Japan
| | - Hidenori Otsuka
- Graduate School of Science and Department of
Applied Chemistry, Faculty of
Science, Tokyo University of Science, 1-3 Kagurazaka,
Shinjuku-ku, Tokyo 162-8601, Japan
- E-mail: . Phone: +81-3-5228-8265. Fax: +81-3-5261-4631 (H.O.)
| |
Collapse
|
47
|
Huynh CT, Liu F, Cheng Y, Coughlin KA, Alsberg E. Thiol-Epoxy "Click" Chemistry to Engineer Cytocompatible PEG-Based Hydrogel for siRNA-Mediated Osteogenesis of hMSCs. ACS APPLIED MATERIALS & INTERFACES 2018; 10:25936-25942. [PMID: 29986132 PMCID: PMC6930143 DOI: 10.1021/acsami.8b07167] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Thiol-epoxy "click" chemistry is employed for the first time to engineer a new cytocompatible PEG-based hydrogel system in aqueous media with the ability to encapsulate human mesenchymal stem cells (hMSCs) and control their fate for tissue regeneration. Cells were easily encapsulated into the hydrogels and exhibited high cell viability over 4 weeks of culture regardless of the presence of siRNA, complexed with polyethylenimine (PEI) in the form of siRNA/PEI nanocomplexes, indicating the biocompatibility of the developed hydrogel. Loading pro-osteogenic siNoggin in the hydrogel significantly enhanced the osteogenesis of encapsulated hMSCs, demonstrating the potential application of this system in tissue engineering.
Collapse
Affiliation(s)
- Cong Truc Huynh
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio 44106, United States
| | - Fangze Liu
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio 44106, United States
| | - Yuxuan Cheng
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio 44106, United States
| | - Katherine A. Coughlin
- Department of Biomedical Engineering, Northwestern University, Evanston, Illinois 60208, United States
| | - Eben Alsberg
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio 44106, United States
- Department of Orthopaedic Surgery, Case Western Reserve University, Cleveland, Ohio 44106, United States
| |
Collapse
|
48
|
Grier WK, Tiffany AS, Ramsey MD, Harley BA. Incorporating β-cyclodextrin into collagen scaffolds to sequester growth factors and modulate mesenchymal stem cell activity. Acta Biomater 2018; 76:116-125. [PMID: 29944975 DOI: 10.1016/j.actbio.2018.06.033] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Revised: 06/21/2018] [Accepted: 06/22/2018] [Indexed: 12/31/2022]
Abstract
The development of biomaterials for a range of tissue engineering applications increasingly requires control over the bioavailability of biomolecular cues such as growth factors in order to promote desired cell responses. While efforts have predominantly concentrated on covalently-bound or freely-diffusible incorporation of biomolecules in porous, three-dimensional biomaterials, opportunities exist to exploit transient interactions to concentrate growth factor activity over desired time frames. Here, we report the incorporation of β-cyclodextrin into a model collagen-GAG scaffold as a means to exploit the passive sequestration and release of growth factors via guest-host interactions to control mesenchymal stem cell differentiation. Collagen-GAG scaffolds that incorporate β-cyclodextrin show improved sequestration as well as extended retention and release of TGF-β1. We further show extended retention and release of TGF-β1 and BMP-2 from β-cyclodextrin modified scaffolds was sufficient to influence the metabolic activity and proliferation of mesenchymal stem cells as well as differential activation of Smad 2/3 and Smad 1/5/8 pathways associated with differential osteo-chondral differentiation. Further, gene expression analysis showed TGF-β1 release from β-cyclodextrin CG scaffolds promoted early chondrogenic-specific differentiation. Ultimately, this work establishes a novel method for the incorporation and display of growth factors within CG scaffolds via supramolecular interactions. Such a design framework offers opportunities to selectively alter the bioavailability of multiple biomolecules within a three-dimensional collagen-GAG scaffold to enhance cell activity for a range of musculoskeletal regenerative medicine applications. STATEMENT OF SIGNIFICANCE We describe the incorporation of β-cyclodextrin into a model CG-scaffold under development for musculoskeletal tissue engineering applications. We show β-cyclodextrin modified scaffolds promote the sequestration of soluble TGF-β1 and BMP-2 via guest-host interactions, leading to extended retention and release. Further, β-cyclodextrin modified CG scaffolds promote TGF-β1 or BMP-2 specific Smad signaling pathway activation associated with divergent osseous versus chondrogenic differentiation pathways in mesenchymal stem cells.
Collapse
|
49
|
Maji S, Agarwal T, Das J, Maiti TK. Development of gelatin/carboxymethyl chitosan/nano-hydroxyapatite composite 3D macroporous scaffold for bone tissue engineering applications. Carbohydr Polym 2018; 189:115-125. [PMID: 29580388 DOI: 10.1016/j.carbpol.2018.01.104] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Revised: 01/10/2018] [Accepted: 01/31/2018] [Indexed: 12/26/2022]
Abstract
The present study delineates a relatively simpler approach for fabrication of a macroporous three-dimensional scaffold for bone tissue engineering. The novelty of the work is to obtain a scaffold with macroporosity (interconnected networks) through a combined approach of high stirring induced foaming of the gelatin/carboxymethyl chitosan (CMC)/nano-hydroxyapatite (nHAp) matrix followed by freeze drying. The fabricated macroporous (SGC) scaffold had a greater pore size, higher porosity, higher water retention capacity, slow and sustained enzymatic degradation rate along with higher compressive strength compared to that of non-macroporous (NGC, prepared by conventional freeze drying methodology) scaffold. The biological studies revealed the increased percentage of viability, proliferation, and differentiation as well as higher mineralization of differentiated human Wharton's jelly MSC microtissue (wjhMSC-MT) on SGC as compared to NGC scaffold. RT-PCR also showed enhanced expression level of collagen type I, osteocalcin and Runx2 when seeded on SGC. μCT and histological analysis further revealed a penetration of cellular spheroid to a greater depth in SGC scaffold than NGC scaffold. Furthermore, the effect of cryopreservation on microtissue survival on the three-dimensional construct revealed significant higher viability upon revival in macroporous SGC scaffolds. These results together suggest that high stirring based macroporous scaffolds could have a potential application in bone tissue engineering.
Collapse
Affiliation(s)
- Somnath Maji
- Department of Biotechnology, Indian Institute of Technology, Kharagpur-721302, West Bengal, India.
| | - Tarun Agarwal
- Department of Biotechnology, Indian Institute of Technology, Kharagpur-721302, West Bengal, India.
| | - Joyjyoti Das
- Department of Biotechnology, Indian Institute of Technology, Kharagpur-721302, West Bengal, India.
| | - Tapas Kumar Maiti
- Department of Biotechnology, Indian Institute of Technology, Kharagpur-721302, West Bengal, India.
| |
Collapse
|
50
|
Müller WEG, Neufurth M, Wang S, Ackermann M, Muñoz-Espí R, Feng Q, Lu Q, Schröder HC, Wang X. Amorphous, Smart, and Bioinspired Polyphosphate Nano/Microparticles: A Biomaterial for Regeneration and Repair of Osteo-Articular Impairments In-Situ. Int J Mol Sci 2018; 19:E427. [PMID: 29385104 PMCID: PMC5855649 DOI: 10.3390/ijms19020427] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Revised: 01/27/2018] [Accepted: 01/29/2018] [Indexed: 12/19/2022] Open
Abstract
Using femur explants from mice as an in vitro model, we investigated the effect of the physiological polymer, inorganic polyphosphate (polyP), on differentiation of the cells of the bone marrow in their natural microenvironment into the osteogenic and chondrogenic lineages. In the form of amorphous Ca-polyP nano/microparticles, polyP retains its function to act as both an intra- and extracellular metabolic fuel and a stimulus eliciting morphogenetic signals. The method for synthesis of the nano/microparticles with the polyanionic polyP also allowed the fabrication of hybrid particles with the bisphosphonate zoledronic acid, a drug used in therapy of bone metastases in cancer patients. The results revealed that the amorphous Ca-polyP particles promote the growth/viability of mesenchymal stem cells, as well as the osteogenic and chondrogenic differentiation of the bone marrow cells in rat femur explants, as revealed by an upregulation of the expression of the transcription factors SOX9 (differentiation towards osteoblasts) and RUNX2 (chondrocyte differentiation). In parallel to this bone anabolic effect, incubation of the femur explants with these particles significantly reduced the expression of the gene encoding the osteoclast bone-catabolic enzyme, cathepsin-K, while the expression of the tartrate-resistant acid phosphatase remained unaffected. The gene expression data were supported by the finding of an increased mineralization of the cells in the femur explants in response to the Ca-polyP particles. Finally, we show that the hybrid particles of polyP complexed with zoledronic acid exhibit both the cytotoxic effect of the bisphosphonate and the morphogenetic and mineralization inducing activity of polyP. Our results suggest that the Ca-polyP nano/microparticles are not only a promising scaffold material for repairing long bone osteo-articular damages but can also be applied, as a hybrid with zoledronic acid, as a drug delivery system for treatment of bone metastases. The polyP particles are highlighted as genuine, smart, bioinspired nano/micro biomaterials.
Collapse
Affiliation(s)
- Werner E G Müller
- ERC Advanced Investigator Grant Research Group at the Institute for Physiological Chemistry, University Medical Center of the Johannes Gutenberg University Mainz, Duesbergweg 6, 55128 Mainz, Germany.
| | - Meik Neufurth
- ERC Advanced Investigator Grant Research Group at the Institute for Physiological Chemistry, University Medical Center of the Johannes Gutenberg University Mainz, Duesbergweg 6, 55128 Mainz, Germany.
| | - Shunfeng Wang
- ERC Advanced Investigator Grant Research Group at the Institute for Physiological Chemistry, University Medical Center of the Johannes Gutenberg University Mainz, Duesbergweg 6, 55128 Mainz, Germany.
| | - Maximilian Ackermann
- Institute of Functional and Clinical Anatomy, University Medical Center of the Johannes Gutenberg University Mainz, Johann Joachim Becher Weg 13, 55099 Mainz, Germany.
| | - Rafael Muñoz-Espí
- Institute of Materials Science (ICMUV), Universitat de València, C/Catedràtic José Beltrán 2, Paterna, 46980 València, Spain.
| | - Qingling Feng
- Key Laboratory of Advanced Materials of Ministry of Education of China, School of Materials Science and Engineering, Tsinghua University, Beijing 100084, China.
| | - Qiang Lu
- ERC Advanced Investigator Grant Research Group at the Institute for Physiological Chemistry, University Medical Center of the Johannes Gutenberg University Mainz, Duesbergweg 6, 55128 Mainz, Germany.
| | - Heinz C Schröder
- ERC Advanced Investigator Grant Research Group at the Institute for Physiological Chemistry, University Medical Center of the Johannes Gutenberg University Mainz, Duesbergweg 6, 55128 Mainz, Germany.
| | - Xiaohong Wang
- ERC Advanced Investigator Grant Research Group at the Institute for Physiological Chemistry, University Medical Center of the Johannes Gutenberg University Mainz, Duesbergweg 6, 55128 Mainz, Germany.
| |
Collapse
|