1
|
Salehi S, Brambilla S, Rasponi M, Lopa S, Moretti M. Development of a Microfluidic Vascularized Osteochondral Model as a Drug Testing Platform for Osteoarthritis. Adv Healthc Mater 2024; 13:e2402350. [PMID: 39370575 DOI: 10.1002/adhm.202402350] [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: 06/28/2024] [Revised: 09/17/2024] [Indexed: 10/08/2024]
Abstract
Osteoarthritis (OA) is a degenerative joint disease characterized by changes in cartilage and subchondral bone. To date, there are no available drugs that can counteract the progression of OA, partly due to the inadequacy of current models to recapitulate the relevant cellular complexity. In this study, an osteochondral microfluidic model is developed using human primary cells to mimic an OA-like microenvironment and this study validates it as a drug testing platform. In the model, the cartilage compartment is created by embedding articular chondrocytes in fibrin hydrogel while the bone compartment is obtained by embedding osteoblasts, osteoclasts, endothelial cells, and mesenchymal stem cells in a fibrin hydrogel enriched with calcium phosphate nanoparticles. After developing and characterizing the model, Interleukin-1β is applied to induce OA-like conditions. Subsequently, the model potential is evaluated as a drug testing platform by assessing the effect of two anti-inflammatory drugs (Interleukin-1 Receptor antagonist and Celecoxib) on the regulation of inflammation- and matrix degradation-related markers. The model responded to inflammation and demonstrated differences in drug efficacy. Finally, it compares the behavior of the "Cartilage" and "Cartilage+Bone" models, emphasizing the necessity of incorporating both cartilage and bone compartments to capture the complex pathophysiology of OA.
Collapse
Affiliation(s)
- Shima Salehi
- Cell and Tissue Engineering Laboratory, IRCCS Istituto Ortopedico Galeazzi, Via Belgioioso 173, Milan, 20157, Italy
| | - Stefania Brambilla
- Cell and Tissue Engineering Laboratory, IRCCS Istituto Ortopedico Galeazzi, Via Belgioioso 173, Milan, 20157, Italy
| | - Marco Rasponi
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Via Ponzio 34/5, Milan, 20133, Italy
| | - Silvia Lopa
- Cell and Tissue Engineering Laboratory, IRCCS Istituto Ortopedico Galeazzi, Via Belgioioso 173, Milan, 20157, Italy
| | - Matteo Moretti
- Cell and Tissue Engineering Laboratory, IRCCS Istituto Ortopedico Galeazzi, Via Belgioioso 173, Milan, 20157, Italy
- Regenerative Medicine Technologies Lab, Laboratories for Translational Research (LRT), Ente Ospedaliero Cantonale (EOC), Via Chiesa 5, Bellinzona, 6500, Switzerland
- Service of Orthopaedics and Traumatology, Department of Surgery, EOC, Via Tesserete 46, Lugano, 6900, Switzerland
- Euler Institute, Faculty of Biomedical Sciences, Università della Svizzera Italiana (USI), Via Buffi 13, Lugano, 6900, Switzerland
| |
Collapse
|
2
|
Deng L, Liu Y, Wu Q, Lai S, Yang Q, Mu Y, Dong M. Exosomes to exosome-functionalized scaffolds: a novel approach to stimulate bone regeneration. Stem Cell Res Ther 2024; 15:407. [PMID: 39521993 PMCID: PMC11550564 DOI: 10.1186/s13287-024-04024-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024] Open
Abstract
Bone regeneration is a complex biological process that relies on the orchestrated interplay of various cellular and molecular events. Bone tissue engineering is currently the most promising method for treating bone regeneration. However, the immunogenicity, stable and cell quantity of seed cells limited their application. Recently, exosomes, which are small extracellular vesicles released by cells, have been found to effectively address these problems and better induce bone regeneration. Meanwhile, a growing line of research has shown the cargos of exosomes may provide effective therapeutic and biomarker tools for bone repair, including miRNA, lncRNA, and proteins. Moreover, engineered scaffolds loaded with exosomes can offer a cell-free bone repair strategy, addressing immunogenicity concerns and providing a more stable functional performance. Herein, we provide a comprehensive summary of the role played by scaffolds loaded with exosomes in bone regeneration, drawing on a systematic analysis of relevant literature available on PubMed, Scopus, and Google Scholar database.
Collapse
Affiliation(s)
- Li Deng
- Center for Medicine Research and Translation, Chengdu Fifth People's Hospital (The Second Clinical Medical College, Affiliated Fifth People's Hospital of Chengdu University of Traditional Chinese Medicine), Chengdu, 611135, Sichuan, China
| | - Yang Liu
- Center for Medicine Research and Translation, Chengdu Fifth People's Hospital (The Second Clinical Medical College, Affiliated Fifth People's Hospital of Chengdu University of Traditional Chinese Medicine), Chengdu, 611135, Sichuan, China
| | - Qian Wu
- Center for Medicine Research and Translation, Chengdu Fifth People's Hospital (The Second Clinical Medical College, Affiliated Fifth People's Hospital of Chengdu University of Traditional Chinese Medicine), Chengdu, 611135, Sichuan, China
| | - Shuang Lai
- Stomatology Department, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 611731, China
| | - Qiu Yang
- Center for Medicine Research and Translation, Chengdu Fifth People's Hospital (The Second Clinical Medical College, Affiliated Fifth People's Hospital of Chengdu University of Traditional Chinese Medicine), Chengdu, 611135, Sichuan, China
| | - Yandong Mu
- Stomatology Department, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 611731, China.
| | - Mingqing Dong
- Center for Medicine Research and Translation, Chengdu Fifth People's Hospital (The Second Clinical Medical College, Affiliated Fifth People's Hospital of Chengdu University of Traditional Chinese Medicine), Chengdu, 611135, Sichuan, China.
| |
Collapse
|
3
|
Zhang B, Berilla J, Cho S, Somoza RA, Welter JF, Alexander PE, Baskaran H. Synergistic effects of biological stimuli and flexion induce microcavities promote hypertrophy and inhibit chondrogenesis during in vitro culture of human mesenchymal stem cell aggregates. Biotechnol J 2024; 19:e2400060. [PMID: 39295570 DOI: 10.1002/biot.202400060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 06/26/2024] [Accepted: 07/30/2024] [Indexed: 09/21/2024]
Abstract
Interzone/cavitation are key steps in early stage joint formation that have not been successfully developed in vitro. Further, current models of endochondral ossification, an important step in early bone formation, lack key morphology morphological structures such as microcavities found during development in vivo. This is possibly due to the lack of appropriate strategies for incorporating chemical and mechanical stimuli that are thought to be involved in joint development. We designed a bioreactor system and investigated the synergic effect of chemical stimuli (chondrogenesis-inducing [CIM] and hypertrophy-inducing medium [HIM]) and mechanical stimuli (flexion) on the growth of human mesenchymal stem cells (hMSCs) based linear aggregates under different conditions over 4 weeks of perfusion culture. Computational studies were used to evaluate tissue stress qualitatively. After harvesting, both Safranin-O and hematoxylin & eosin (H&E) staining histology demonstrated microcavity structures and void structures in the region of higher stresses for tissue aggregates cultured only in HIM under flexion. In comparison to either HIM treatment or flexion only, increased glycosaminoglycan (GAG) content in the extracellular matrix (ECM) at this region indicates the morphological change resembles the early stage of joint cavitation; while decreased type II collagen (Col II), and increased type X collagen (Col X) and vascular endothelial growth factor (VEGF) with a clear boundary in the staining section indicates it resembles the early stage of ossification. Further, cell alignment analysis indicated that cells were mostly oriented toward the direction of flexion in high-stress region only in HIM under flexion, resembling cell morphology in both joint cavitation and hypertrophic cartilage in growth plate. Collectively, our results suggest that flexion and HIM inhibit chondrogenesis and promote hypertrophy and development of microcavities that resemble the early stage of joint cavitation and endochondral ossification. We believe the tissue model described in this work can be used to develop in vitro models of joint tissue for applications such as pathophysiology and drug discovery.
Collapse
Affiliation(s)
- Bo Zhang
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, USA
| | - Jim Berilla
- Case School of Engineering, Case Western Reserve University, Cleveland, Ohio, USA
| | - Sungwoo Cho
- Department of Biochemistry, Case Western Reserve University, Cleveland, Ohio, USA
| | - Rodrigo A Somoza
- Department of Biology, Case Western Reserve University, Cleveland, Ohio, USA
| | - Jean F Welter
- Department of Biology, Case Western Reserve University, Cleveland, Ohio, USA
| | - Peter E Alexander
- Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Harihara Baskaran
- Department of Chemical and Biomolecular Engineering, Case Western Reserve University, Cleveland, Ohio, USA
| |
Collapse
|
4
|
Lau CS, Park SY, Ethiraj LP, Singh P, Raj G, Quek J, Prasadh S, Choo Y, Goh BT. Role of Adipose-Derived Mesenchymal Stem Cells in Bone Regeneration. Int J Mol Sci 2024; 25:6805. [PMID: 38928517 PMCID: PMC11204188 DOI: 10.3390/ijms25126805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 06/13/2024] [Accepted: 06/14/2024] [Indexed: 06/28/2024] Open
Abstract
Bone regeneration involves multiple factors such as tissue interactions, an inflammatory response, and vessel formation. In the event of diseases, old age, lifestyle, or trauma, bone regeneration can be impaired which could result in a prolonged healing duration or requiring an external intervention for repair. Currently, bone grafts hold the golden standard for bone regeneration. However, several limitations hinder its clinical applications, e.g., donor site morbidity, an insufficient tissue volume, and uncertain post-operative outcomes. Bone tissue engineering, involving stem cells seeded onto scaffolds, has thus been a promising treatment alternative for bone regeneration. Adipose-derived mesenchymal stem cells (AD-MSCs) are known to hold therapeutic value for the treatment of various clinical conditions and have displayed feasibility and significant effectiveness due to their ease of isolation, non-invasive, abundance in quantity, and osteogenic capacity. Notably, in vitro studies showed AD-MSCs holding a high proliferation capacity, multi-differentiation potential through the release of a variety of factors, and extracellular vesicles, allowing them to repair damaged tissues. In vivo and clinical studies showed AD-MSCs favoring better vascularization and the integration of the scaffolds, while the presence of scaffolds has enhanced the osteogenesis potential of AD-MSCs, thus yielding optimal bone formation outcomes. Effective bone regeneration requires the interplay of both AD-MSCs and scaffolds (material, pore size) to improve the osteogenic and vasculogenic capacity. This review presents the advances and applications of AD-MSCs for bone regeneration and bone tissue engineering, focusing on the in vitro, in vivo, and clinical studies involving AD-MSCs for bone tissue engineering.
Collapse
Affiliation(s)
- Chau Sang Lau
- National Dental Centre Singapore, National Dental Research Institute Singapore, Singapore 168938, Singapore; (C.S.L.); (S.Y.P.); (L.P.E.); (G.R.)
- Oral Health Academic Clinical Programme, Duke-NUS Medical School, Singapore 169857, Singapore
| | - So Yeon Park
- National Dental Centre Singapore, National Dental Research Institute Singapore, Singapore 168938, Singapore; (C.S.L.); (S.Y.P.); (L.P.E.); (G.R.)
| | - Lalith Prabha Ethiraj
- National Dental Centre Singapore, National Dental Research Institute Singapore, Singapore 168938, Singapore; (C.S.L.); (S.Y.P.); (L.P.E.); (G.R.)
- Oral Health Academic Clinical Programme, Duke-NUS Medical School, Singapore 169857, Singapore
| | - Priti Singh
- National Dental Centre Singapore, National Dental Research Institute Singapore, Singapore 168938, Singapore; (C.S.L.); (S.Y.P.); (L.P.E.); (G.R.)
| | - Grace Raj
- National Dental Centre Singapore, National Dental Research Institute Singapore, Singapore 168938, Singapore; (C.S.L.); (S.Y.P.); (L.P.E.); (G.R.)
| | - Jolene Quek
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore; (J.Q.); (Y.C.)
| | - Somasundaram Prasadh
- Center for Clean Energy Engineering, University of Connecticut, Storrs, CT 06269, USA;
| | - Yen Choo
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore; (J.Q.); (Y.C.)
| | - Bee Tin Goh
- National Dental Centre Singapore, National Dental Research Institute Singapore, Singapore 168938, Singapore; (C.S.L.); (S.Y.P.); (L.P.E.); (G.R.)
- Oral Health Academic Clinical Programme, Duke-NUS Medical School, Singapore 169857, Singapore
| |
Collapse
|
5
|
Li X, Zhu L, Che Z, Liu T, Yang C, Huang L. Progress of research on the surface functionalization of tantalum and porous tantalum in bone tissue engineering. Biomed Mater 2024; 19:042009. [PMID: 38838694 DOI: 10.1088/1748-605x/ad5481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Accepted: 06/05/2024] [Indexed: 06/07/2024]
Abstract
Tantalum and porous tantalum are ideal materials for making orthopedic implants due to their stable chemical properties and excellent biocompatibility. However, their utilization is still affected by loosening, infection, and peripheral inflammatory reactions, which sometimes ultimately lead to implant removal. An ideal bone implant should have exceptional biological activity, which can improve the surrounding biological microenvironment to enhance bone repair. Recent advances in surface functionalization have produced various strategies for developing compatibility between either of the two materials and their respective microenvironments. This review provides a systematic overview of state-of-the-art strategies for conferring biological functions to tantalum and porous tantalum implants. Furthermore, the review describes methods for preparing active surfaces and different bioactive substances that are used, summarizing their functions. Finally, this review discusses current challenges in the development of optimal bone implant materials.
Collapse
Affiliation(s)
- Xudong Li
- The Second Hospital of Jilin University, Changchun 130041, People's Republic of China
| | - Liwei Zhu
- The Second Hospital of Jilin University, Changchun 130041, People's Republic of China
| | - Zhenjia Che
- The Second Hospital of Jilin University, Changchun 130041, People's Republic of China
| | - Tengyue Liu
- The Second Hospital of Jilin University, Changchun 130041, People's Republic of China
| | - Chengzhe Yang
- The Second Hospital of Jilin University, Changchun 130041, People's Republic of China
| | - Lanfeng Huang
- The Second Hospital of Jilin University, Changchun 130041, People's Republic of China
| |
Collapse
|
6
|
Bastos AR, da Silva LP, Maia FR, Franco A, Noro J, Silva C, Oliveira JM, Reis RL, Correlo VM. Hydroxyapatite/alginate/gellan gum inks with osteoconduction and osteogenic potential for bioprinting bone tissue analogues. Int J Biol Macromol 2024; 271:132611. [PMID: 38797304 DOI: 10.1016/j.ijbiomac.2024.132611] [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: 02/20/2024] [Revised: 05/10/2024] [Accepted: 05/22/2024] [Indexed: 05/29/2024]
Abstract
There is a growing demand for engineered bone tissues custom-designed to match the patient-specific defect size and in vitro models for studying bone diseases and/or drug screening. Herein, we propose a bioprinted bone tissue construct using SaOs-2 cells within alginate/gellan gum/hydroxyapatite inks. Different ink formulations were developed with varying hydroxyapatite content and then evaluated for viscoelasticity, printability, biomineralization properties, post-printing viability, proliferation, metabolic activity, and osteogenic phenotype of SaOs-2-encapsulated cells. Results indicate that ink formulations exhibit non-Newtonian shear-thinning behaviour, maintaining shape integrity and structural stability post-printing. Ink mineralization rates increase with the hydroxyapatite content, rendering them suitable for bone defect strategies. Post-printed cells in the developed constructs remain live, spreading, and metabolically active but do not proliferate. Osteogenic gene and protein expression, both early and late, show upregulation at day 7 relative to day 1, followed by downregulation at day 14. Lower hydroxyapatite content inks demonstrate up to fourfold upregulation in genes and proteins at most time points. Additionally, these constructs release calcium and phosphate at levels conducive to mineralization. Overall, the tissue-engineered miniaturized constructs not only meet the criteria for early-stage bone defect/fracture regeneration but also serve as a promising platform for drug screening and evaluating potential therapeutic treatments.
Collapse
Affiliation(s)
- Ana Raquel Bastos
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal; ICVS/3B's - PT Government Associated Laboratory, Portugal
| | - Lucília P da Silva
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal; ICVS/3B's - PT Government Associated Laboratory, Portugal.
| | - F Raquel Maia
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal; ICVS/3B's - PT Government Associated Laboratory, Portugal
| | - Albina Franco
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal; ICVS/3B's - PT Government Associated Laboratory, Portugal
| | - Jennifer Noro
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal; ICVS/3B's - PT Government Associated Laboratory, Portugal
| | - Carla Silva
- Center of Biological Engineering, University of Minho, Braga, Portugal
| | - J Miguel Oliveira
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal; ICVS/3B's - PT Government Associated Laboratory, Portugal
| | - Rui Luís Reis
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal; ICVS/3B's - PT Government Associated Laboratory, Portugal
| | - Vitor Manuel Correlo
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal; ICVS/3B's - PT Government Associated Laboratory, Portugal.
| |
Collapse
|
7
|
Xu Z, Wang B, Huang R, Guo M, Han D, Yin L, Zhang X, Huang Y, Li X. Efforts to promote osteogenesis-angiogenesis coupling for bone tissue engineering. Biomater Sci 2024; 12:2801-2830. [PMID: 38683241 DOI: 10.1039/d3bm02017g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/01/2024]
Abstract
Repair of bone defects exceeding a critical size has been always a big challenge in clinical practice. Tissue engineering has exhibited great potential to effectively repair the defects with less adverse effect than traditional bone grafts, during which how to induce vascularized bone formation has been recognized as a critical issue. Therefore, recently many studies have been launched to attempt to promote osteogenesis-angiogenesis coupling. This review summarized comprehensively and explored in depth current efforts to ameliorate the coupling of osteogenesis and angiogenesis from four aspects, namely the optimization of scaffold components, modification of scaffold structures, loading strategies for bioactive substances, and employment tricks for appropriate cells. Especially, the advantages and the possible reasons for every strategy, as well as the challenges, were elaborated. Furthermore, some promising research directions were proposed based on an in-depth analysis of the current research. This paper will hopefully spark new ideas and approaches for more efficiently boosting new vascularized bone formations.
Collapse
Affiliation(s)
- Zhiwei Xu
- College of Lab Medicine, Hebei North University, Zhangjiakou 075000, China
| | - Bingbing Wang
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, 37 Xueyuan Rd, Haidian District, Beijing, 100083, China.
| | - Ruoyu Huang
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, 37 Xueyuan Rd, Haidian District, Beijing, 100083, China.
| | - Mengyao Guo
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, 37 Xueyuan Rd, Haidian District, Beijing, 100083, China.
| | - Di Han
- College of Lab Medicine, Hebei North University, Zhangjiakou 075000, China
| | - Lan Yin
- Key Laboratory of Advanced Materials of Ministry of Education, Tsinghua University, Beijing 100084, China
| | - Xiaoyun Zhang
- College of Lab Medicine, Hebei North University, Zhangjiakou 075000, China
| | - Yong Huang
- College of Lab Medicine, Hebei North University, Zhangjiakou 075000, China
| | - Xiaoming Li
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, 37 Xueyuan Rd, Haidian District, Beijing, 100083, China.
| |
Collapse
|
8
|
A L, Elsen R, Nayak S. Artificial Intelligence-Based 3D Printing Strategies for Bone Scaffold Fabrication and Its Application in Preclinical and Clinical Investigations. ACS Biomater Sci Eng 2024; 10:677-696. [PMID: 38252807 DOI: 10.1021/acsbiomaterials.3c01368] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
3D printing has become increasingly popular in the field of bone tissue engineering. However, the mechanical properties, biocompatibility, and porosity of the 3D printed bone scaffolds are major requirements for tissue regeneration and implantation as well. Designing the scaffold architecture in accordance with the need to create better mechanical and biological stimuli is necessary to achieve unique scaffold properties. To accomplish this, different 3D designing strategies can be utilized with the help of the scaffold design library and artificial intelligence (AI). The implementation of AI to assist the 3D printing process can enable it to predict, adapt, and control the parameters on its own, which lowers the risk of errors. This Review emphasizes 3D design and fabrication of bone scaffold using different materials and the use of AI-aided 3D printing strategies. Also, the adaption of AI to 3D printing helps to develop patient-specific scaffolds based on different requirements, thus providing feedback and adequate data for reproducibility, which can be improvised in the future. These printed scaffolds can also serve as an alternative to preclinical animal test models to cut costs and prevent immunological interference.
Collapse
Affiliation(s)
- Logeshwaran A
- School of Bio Sciences and Technology, Vellore Institute of Technology (VIT), Katpadi, Vellore, Tamil Nadu 632014, India
| | - Renold Elsen
- School of Mechanical Engineering, Vellore Institute of Technology (VIT), Katpadi, Vellore, Tamil Nadu 632014, India
| | - Sunita Nayak
- School of Bio Sciences and Technology, Vellore Institute of Technology (VIT), Katpadi, Vellore, Tamil Nadu 632014, India
| |
Collapse
|
9
|
Duan QY, Zhu YX, Jia HR, Wang SH, Wu FG. Nanogels: Synthesis, properties, and recent biomedical applications. PROGRESS IN MATERIALS SCIENCE 2023; 139:101167. [DOI: 10.1016/j.pmatsci.2023.101167] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
10
|
Bosio VE, Rybner C, Kaplan DL. Concentric-mineralized hybrid silk-based scaffolds for bone tissue engineering in vitro models. J Mater Chem B 2023; 11:7998-8006. [PMID: 37526619 PMCID: PMC10563295 DOI: 10.1039/d3tb00717k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/02/2023]
Abstract
There are many challenges in the development of 3D-tissue models for studying bone physiology and disease. Silk fibroin (SF), a natural fibrous protein used in biomedical applications has been studied for bone tissue engineering (TE) due to its mechanical properties, biocompatibility and biodegradability. However, low osteogenic capacity as well as the necessity to reinforce the protein mechanically for some orthopedic applications prompts the need for further designs for SF-based materials for TE bone. Concentric mineralized porous SF-based scaffolds were developed to improve mechanics and mineralization towards osteoregeneration. Hybrid SF silica microparticles (MP) or calcium carbonate nano-structured microparticles (NMP) were seeded with hMSCs co-cultured under osteogenic and osteoclastic conditions with THP-1 human monocytes up to 10 weeks to simulate and recapitulate bone regeneration. Scaffolds with appropriate pore size for cell infiltration, resulted in improved compressive strength, increased cell attachment and higher levels of expression of osteogenic markers and mineralization after adding the NMPs, compared to controls systems without these particles. These hybrid SF-based 3D-structures can provide improved scaffold designs for in vitro bone TE.
Collapse
Affiliation(s)
- Valeria E Bosio
- BIOMIT Lab (Biomaterials in Tissue Engineering Lab) Institute of Physics La Plata (IFLP), University of La Plata & CONICET, Diag. 113 e/63 y 64, CP 1900, La Plata, Buenos Aires, Argentina.
- Department of Biomaterials, Celll Institute, Ciudad de Buenos Aires, Argentina
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts 02155, USA
| | - Christofer Rybner
- BIOMIT Lab (Biomaterials in Tissue Engineering Lab) Institute of Physics La Plata (IFLP), University of La Plata & CONICET, Diag. 113 e/63 y 64, CP 1900, La Plata, Buenos Aires, Argentina.
- Department of Biomaterials, Celll Institute, Ciudad de Buenos Aires, Argentina
| | - David L Kaplan
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts 02155, USA
| |
Collapse
|
11
|
Kazimierczak P, Kalisz G, Sroka-Bartnicka A, Przekora A. Effectiveness of the production of tissue-engineered living bone graft: a comparative study using perfusion and rotating bioreactor systems. Sci Rep 2023; 13:13737. [PMID: 37612367 PMCID: PMC10447456 DOI: 10.1038/s41598-023-41003-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 08/20/2023] [Indexed: 08/25/2023] Open
Abstract
Bioreactor systems are very precious tools to generate living bone grafts in vitro. The aim of this study was to compare the effectiveness of rotating and perfusion bioreactor in the production of a living bone construct. Human bone marrow-derived mesenchymal stem cells (BMDSCs) were seeded on the surfaces of hydroxyapatite-based scaffolds and cultured for 21 days in three different conditions: (1) static 3D culture, (2) 3D culture in a perfusion bioreactor, and (3) dynamic 3D culture in a rotating bioreactor. Quantitative evaluation of cell number showed that cultivation in the perfusion bioreactor significantly reduced cell proliferation compared to the rotating bioreactor and static culture. Osteogenic differentiation test demonstrated that BMDSCs cultured in the rotating bioreactor produced significantly greater amount of osteopontin compared to the cells cultured in the perfusion bioreactor. Moreover, Raman spectroscopy showed that cultivation of BMDSCs in the rotating bioreactor enhanced extracellular matrix (ECM) mineralization that was characterized by B-type carbonated substitution of hydroxyapatite (associated with PO43- groups) and higher mineral-to-matrix ratio compared to the ECM of cells cultured in the perfusion system. Thus, it was concluded that the rotating bioreactor was much more effective than the perfusion one in the generation of bone tissue construct in vitro.
Collapse
Affiliation(s)
- Paulina Kazimierczak
- Independent Unit of Tissue Engineering and Regenerative Medicine, Medical University of Lublin, Chodzki 1 Street, 20-093, Lublin, Poland.
| | - Grzegorz Kalisz
- Independent Unit of Spectroscopy and Chemical Imaging, Medical University of Lublin, Chodzki 4a Street, 20-093, Lublin, Poland
| | - Anna Sroka-Bartnicka
- Independent Unit of Spectroscopy and Chemical Imaging, Medical University of Lublin, Chodzki 4a Street, 20-093, Lublin, Poland
| | - Agata Przekora
- Independent Unit of Tissue Engineering and Regenerative Medicine, Medical University of Lublin, Chodzki 1 Street, 20-093, Lublin, Poland
| |
Collapse
|
12
|
Alamán‐Díez P, García‐Gareta E, Arruebo M, Pérez MÁ. A bone-on-a-chip collagen hydrogel-based model using pre-differentiated adipose-derived stem cells for personalized bone tissue engineering. J Biomed Mater Res A 2023; 111:88-105. [PMID: 36205241 PMCID: PMC9828068 DOI: 10.1002/jbm.a.37448] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 08/24/2022] [Accepted: 09/13/2022] [Indexed: 01/12/2023]
Abstract
Mesenchymal stem cells have contributed to the continuous progress of tissue engineering and regenerative medicine. Adipose-derived stem cells (ADSC) possess many advantages compared to other origins including easy tissue harvesting, self-renewal potential, and fast population doubling time. As multipotent cells, they can differentiate into osteoblastic cell linages. In vitro bone models are needed to carry out an initial safety assessment in the study of novel bone regeneration therapies. We hypothesized that 3D bone-on-a-chip models containing ADSC could closely recreate the physiological bone microenvironment and promote differentiation. They represent an intermedium step between traditional 2D-in vitro and in vivo experiments facilitating the screening of therapeutic molecules while saving resources. Herein, we have differentiated ADSC for 7 and 14 days and used them to fabricate in vitro bone models by embedding the pre-differentiated cells in a 3D collagen matrix placed in a microfluidic chip. Osteogenic markers such as alkaline phosphatase activity, calcium mineralization, changes on cell morphology, and expression of specific proteins (bone sialoprotein 2, dentin matrix acidic phosphoprotein-1, and osteocalcin) were evaluated to determine cell differentiation potential and evolution. This is the first miniaturized 3D-in vitro bone model created from pre-differentiated ADSC embedded in a hydrogel collagen matrix which could be used for personalized bone tissue engineering.
Collapse
Affiliation(s)
- Pilar Alamán‐Díez
- Multiscale in Mechanical and Biological Engineering, Aragón Institute of Engineering Research (I3A), Aragón Institute of Healthcare Research (IIS Aragón)University of ZaragozaZaragozaSpain
| | - Elena García‐Gareta
- Multiscale in Mechanical and Biological Engineering, Aragón Institute of Engineering Research (I3A), Aragón Institute of Healthcare Research (IIS Aragón)University of ZaragozaZaragozaSpain,Division of Biomaterials and Tissue Engineering, UCL Eastman Dental InstituteUniversity College LondonLondonUK
| | - Manuel Arruebo
- Aragón Institute of Nanoscience and Materials (INMA), Consejo Superior de Investigaciones Científicas (CSIC)University of ZaragozaZaragozaSpain,Department of Chemical EngineeringUniversity of ZaragozaZaragozaSpain
| | - María Ángeles Pérez
- Multiscale in Mechanical and Biological Engineering, Aragón Institute of Engineering Research (I3A), Aragón Institute of Healthcare Research (IIS Aragón)University of ZaragozaZaragozaSpain
| |
Collapse
|
13
|
Kumar A, Sood A, Singhmar R, Mishra YK, Thakur VK, Han SS. Manufacturing functional hydrogels for inducing angiogenic-osteogenic coupled progressions in hard tissue repairs: prospects and challenges. Biomater Sci 2022; 10:5472-5497. [PMID: 35994005 DOI: 10.1039/d2bm00894g] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
In large bone defects, inadequate vascularization within the engineered constructs has been a major challenge in developing clinically impactful products. It is fairly determined that bone tissues and blood vessels are established concurrently throughout tissue repairs after an injury. Thus, the coupling of angiogenesis-osteogenesis is an essential course of action in bone tissue restoration. The manufacture of biomaterial-based scaffolds plays a decisive role in stimulating angiogenic and osteogenic progressions (instruction of neovascularization and bone mineralization). Bone hydrogels with optimal conditions are more efficient at healing bone defects. There has been a remarkable advancement in producing bone substitutes in the tissue engineering area, but the sufficient and timely vascularization of engineered constructs for optimal tissue integration and regeneration is lacking due to mismatch in the scaffold characteristics and new bone tissue reconstruction. Therefore, various key challenges remain to be overcome. A deep understanding of angiogenesis and osteogenesis progressions is required to manufacture bone hydrogels with satisfactory results. The current review briefly discusses the fundamentals of bone tissues, the significance of angiogenesis-osteogenesis progressions and their inducers, the efficacy of biomaterials and composite hydrogel-promoted neo-vasculogenesis (i.e. angiogenesis) and bone mineralization (i.e. osteogenesis), and related challenges, including future research directions.
Collapse
Affiliation(s)
- Anuj Kumar
- School of Chemical Engineering, Yeungnam University, 280 Daehak-ro, Gyeongsan 38541, South Korea. .,Research Institute of Cell Culture, Yeungnam University, 280 Daehak-ro, Gyeongsan 38541, South Korea
| | - Ankur Sood
- School of Chemical Engineering, Yeungnam University, 280 Daehak-ro, Gyeongsan 38541, South Korea.
| | - Ritu Singhmar
- School of Chemical Engineering, Yeungnam University, 280 Daehak-ro, Gyeongsan 38541, South Korea.
| | - Yogendra Kumar Mishra
- Smart Materials, NanoSYD, Mads Clausen Institute, University of Southern Denmark, Alsion 2, 6400, Sønderborg, Denmark
| | - Vijay Kumar Thakur
- Biorefining and Advanced Materials Research Center, Scotland's Rural College (SRUC), Kings Buildings, Edinburgh EH9 3JG, UK.,School of Engineering, University of Petroleum and Energy Studies (UPES), Dehradun 248007, Uttarakhand, India
| | - Sung Soo Han
- School of Chemical Engineering, Yeungnam University, 280 Daehak-ro, Gyeongsan 38541, South Korea. .,Research Institute of Cell Culture, Yeungnam University, 280 Daehak-ro, Gyeongsan 38541, South Korea
| |
Collapse
|
14
|
Iafrate L, Benedetti MC, Donsante S, Rosa A, Corsi A, Oreffo ROC, Riminucci M, Ruocco G, Scognamiglio C, Cidonio G. Modelling skeletal pain harnessing tissue engineering. IN VITRO MODELS 2022; 1:289-307. [PMID: 36567849 PMCID: PMC9766883 DOI: 10.1007/s44164-022-00028-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 07/01/2022] [Accepted: 07/04/2022] [Indexed: 12/27/2022]
Abstract
Bone pain typically occurs immediately following skeletal damage with mechanical distortion or rupture of nociceptive fibres. The pain mechanism is also associated with chronic pain conditions where the healing process is impaired. Any load impacting on the area of the fractured bone will stimulate the nociceptive response, necessitating rapid clinical intervention to relieve pain associated with the bone damage and appropriate mitigation of any processes involved with the loss of bone mass, muscle, and mobility and to prevent death. The following review has examined the mechanisms of pain associated with trauma or cancer-related skeletal damage focusing on new approaches for the development of innovative therapeutic interventions. In particular, the review highlights tissue engineering approaches that offer considerable promise in the application of functional biomimetic fabrication of bone and nerve tissues. The strategic combination of bone and nerve tissue engineered models provides significant potential to develop a new class of in vitro platforms, capable of replacing in vivo models and testing the safety and efficacy of novel drug treatments aimed at the resolution of bone-associated pain. To date, the field of bone pain research has centred on animal models, with a paucity of data correlating to the human physiological response. This review explores the evident gap in pain drug development research and suggests a step change in approach to harness tissue engineering technologies to recapitulate the complex pathophysiological environment of the damaged bone tissue enabling evaluation of the associated pain-mimicking mechanism with significant therapeutic potential therein for improved patient quality of life. Graphical abstract Rationale underlying novel drug testing platform development. Pain detected by the central nervous system and following bone fracture cannot be treated or exclusively alleviated using standardised methods. The pain mechanism and specificity/efficacy of pain reduction drugs remain poorly understood. In vivo and ex vivo models are not yet able to recapitulate the various pain events associated with skeletal damage. In vitro models are currently limited by their inability to fully mimic the complex physiological mechanisms at play between nervous and skeletal tissue and any disruption in pathological states. Robust innovative tissue engineering models are needed to better understand pain events and to investigate therapeutic regimes.
Collapse
Affiliation(s)
- Lucia Iafrate
- Center for Life Nano- & Neuro-Science (CLN2S), Istituto Italiano di Tecnologia, Rome, Italy
| | - Maria Cristina Benedetti
- Center for Life Nano- & Neuro-Science (CLN2S), Istituto Italiano di Tecnologia, Rome, Italy
- Department of Biology and Biotechnologies “Charles Darwin”, Sapienza University of Rome, Rome, Italy
| | - Samantha Donsante
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Alessandro Rosa
- Center for Life Nano- & Neuro-Science (CLN2S), Istituto Italiano di Tecnologia, Rome, Italy
- Department of Biology and Biotechnologies “Charles Darwin”, Sapienza University of Rome, Rome, Italy
| | - Alessandro Corsi
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Richard O. C. Oreffo
- Bone and Joint Research Group, Stem Cells and Regeneration, Institute of Developmental Sciences, Centre for Human Development, University of Southampton, Southampton, UK
| | - Mara Riminucci
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Giancarlo Ruocco
- Center for Life Nano- & Neuro-Science (CLN2S), Istituto Italiano di Tecnologia, Rome, Italy
| | - Chiara Scognamiglio
- Center for Life Nano- & Neuro-Science (CLN2S), Istituto Italiano di Tecnologia, Rome, Italy
| | - Gianluca Cidonio
- Center for Life Nano- & Neuro-Science (CLN2S), Istituto Italiano di Tecnologia, Rome, Italy
- Bone and Joint Research Group, Stem Cells and Regeneration, Institute of Developmental Sciences, Centre for Human Development, University of Southampton, Southampton, UK
| |
Collapse
|
15
|
Vitamin B 2 Prevents Glucocorticoid-Caused Damage of Blood Vessels in Osteonecrosis of the Femoral Head. BIOMED RESEARCH INTERNATIONAL 2022; 2022:4006184. [PMID: 35845964 PMCID: PMC9279053 DOI: 10.1155/2022/4006184] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 04/25/2022] [Accepted: 05/10/2022] [Indexed: 12/19/2022]
Abstract
Osteonecrosis of the femoral head (ONFH) is a disorder that can cause collapse of the femoral head. The damage and dysfunction of femoral head microvascular endothelial cells are related to the pathogenesis of glucocorticoid-induced ONFH. Reports suggest that vitamin B2 can promote osteoblast differentiation and prevent low bone mineral density and prevent reperfusion oxidative injury. To explore the effect and possible molecular mechanism of vitamin B2 on the ONFH and Human Umbilical Vein Endothelial Cells (HUVECs), we performed a rat model of ONFH by dexamethasone. The rats were randomly divided into four groups: control group, vitamin B2 group, dexamethasone group, and dexamethasone combined with vitamin B2 treatment group. HUVECs were used to further prove the role and mechanism of vitamin B2 in vitro. In patients, according to immunohistochemical and qRT-PCR of the femoral head, the angiogenic capacity of the ONFH femoral head is compromised. In vivo, it showed that vitamin B2 could inhibit glucocorticoid-induced ONFH-like changes in rats by suppressing cell apoptosis, promoting the regeneration of blood vessels, and increasing bone mass. According to in vitro results, vitamin B2 could induce the migration of HUVECs, enhance the expression of angiogenesis-related factors, and inhibit glucocorticoid-induced apoptosis. The underlying mechanism may be that vitamin B2 activates the PI3K signaling pathway. Vitamin B2 alleviated dexamethasone-induced ONFH, and vitamin B2 could promote the proliferation and migration of HUVECs and inhibit their apoptosis by activating the PI3K/Akt signaling pathway. Vitamin B2 may be a potentially effective treatment for ONFH.
Collapse
|
16
|
Lin Z, Zhang X, Fritch MR, Li Z, Kuang B, Alexander PG, Hao T, Cao G, Tan S, Bruce KK, Lin H. Engineering pre-vascularized bone-like tissue from human mesenchymal stem cells through simulating endochondral ossification. Biomaterials 2022; 283:121451. [DOI: 10.1016/j.biomaterials.2022.121451] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 01/28/2022] [Accepted: 02/27/2022] [Indexed: 01/12/2023]
|
17
|
Wan HY, Shin RLY, Chen JCH, Assunção M, Wang D, Nilsson SK, Tuan RS, Blocki A. Dextran sulfate-amplified extracellular matrix deposition promotes osteogenic differentiation of mesenchymal stem cells. Acta Biomater 2022; 140:163-177. [PMID: 34875356 DOI: 10.1016/j.actbio.2021.11.049] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 11/24/2021] [Accepted: 11/30/2021] [Indexed: 12/21/2022]
Abstract
The development of bone-like tissues in vitro that exhibit key features similar to those in vivo is needed to produce tissue models for drug screening and the study of bone physiology and disease pathogenesis. Extracellular matrix (ECM) is a predominant component of bone in vivo; however, as ECM assembly is sub-optimal in vitro, current bone tissue engineering approaches are limited by an imbalance in ECM-to-cell ratio. We amplified the deposition of osteoblastic ECM by supplementing dextran sulfate (DxS) into osteogenically induced cultures of human mesenchymal stem cells (MSCs). DxS, previously implicated to act as a macromolecular crowder, was recently demonstrated to aggregate and co-precipitate major ECM components, including collagen type I, thereby amplifying its deposition. This effect was re-confirmed for MSC cultures undergoing osteogenic induction, where DxS supplementation augmented collagen type I deposition, accompanied by extracellular osteocalcin accumulation. The resulting differentiated osteoblasts exhibited a more mature osteogenic gene expression profile, indicated by a strong upregulation of the intermediate and late osteogenic markers ALP and OCN, respectively. The associated cellular microenvironment was also enriched in bone morphogenetic protein 2 (BMP-2). Interestingly, the resulting decellularized matrices exhibited the strongest osteo-inductive effects on re-seeded MSCs, promoted cell proliferation, osteogenic marker expression and ECM calcification. Taken together, these findings suggest that DxS-mediated enhancement of osteogenic differentiation by MSCs is mediated by the amplified ECM, which is enriched in osteo-inductive factors. We have thus established a simple and reproducible approach to generate ECM-rich bone-like tissue in vitro with sequestration of osteo-inductive factors. STATEMENT OF SIGNIFICANCE: As extracellular matrix (ECM) assembly is significantly retarded in vitro, the imbalance in ECM-to-cell ratio hampers current in vitro bone tissue engineering approaches in their ability to faithfully resemble their in vivo counterpart. We addressed this limitation by leveraging a poly-electrolyte mediated co-assembly and amplified deposition of ECM during osteogenic differentiation of human mesenchymal stem cells (MSCs). The resulting pericelluar space in culture was enriched in organic and inorganic bone ECM components, as well as osteo-inductive factors, which promoted the differentiation of MSCs towards a more mature osteoblastic phenotype. These findings thus demonstrated a simple and reproducible approach to generate ECM-rich bone-like tissue in vitro with a closer recapitulation of the in vivo tissue niche.
Collapse
|
18
|
Davari N, Bakhtiary N, Khajehmohammadi M, Sarkari S, Tolabi H, Ghorbani F, Ghalandari B. Protein-Based Hydrogels: Promising Materials for Tissue Engineering. Polymers (Basel) 2022; 14:986. [PMID: 35267809 PMCID: PMC8914701 DOI: 10.3390/polym14050986] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 02/19/2022] [Accepted: 02/23/2022] [Indexed: 02/01/2023] Open
Abstract
The successful design of a hydrogel for tissue engineering requires a profound understanding of its constituents' structural and molecular properties, as well as the proper selection of components. If the engineered processes are in line with the procedures that natural materials undergo to achieve the best network structure necessary for the formation of the hydrogel with desired properties, the failure rate of tissue engineering projects will be significantly reduced. In this review, we examine the behavior of proteins as an essential and effective component of hydrogels, and describe the factors that can enhance the protein-based hydrogels' structure. Furthermore, we outline the fabrication route of protein-based hydrogels from protein microstructure and the selection of appropriate materials according to recent research to growth factors, crucial members of the protein family, and their delivery approaches. Finally, the unmet needs and current challenges in developing the ideal biomaterials for protein-based hydrogels are discussed, and emerging strategies in this area are highlighted.
Collapse
Affiliation(s)
- Niyousha Davari
- Department of Life Science Engineering, Faculty of New Sciences and Technologies, University of Tehran, Tehran 143951561, Iran;
| | - Negar Bakhtiary
- Burn Research Center, Iran University of Medical Sciences, Tehran 1449614535, Iran;
- Department of Biomaterials, Faculty of Interdisciplinary Science and Technology, Tarbiat Modares University, Tehran 14115114, Iran
| | - Mehran Khajehmohammadi
- Department of Mechanical Engineering, Faculty of Engineering, Yazd University, Yazd 8174848351, Iran;
- Medical Nanotechnology and Tissue Engineering Research Center, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd 8916877391, Iran
| | - Soulmaz Sarkari
- Department of Biomedical Engineering, Science and Research Branch, Islamic Azad University, Tehran 1477893855, Iran;
| | - Hamidreza Tolabi
- New Technologies Research Center (NTRC), Amirkabir University of Technology, Tehran 158754413, Iran;
- Department of Biomedical Engineering, Amirkabir University of Technology (Tehran Polytechnic), Tehran 158754413, Iran
| | - Farnaz Ghorbani
- Institute of Biomaterials, Department of Material Science and Engineering, University of Erlangen-Nuremberg, Cauerstraße 6, 91058 Erlangen, Germany
| | - Behafarid Ghalandari
- State Key Laboratory of Oncogenes and Related Genes, Institute for Personalized Medicine, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| |
Collapse
|
19
|
Zhao Z, Wang M, Shao F, Liu G, Li J, Wei X, Zhang X, Yang J, Cao F, Wang Q, Wang H, Zhao D. Porous tantalum-composited gelatin nanoparticles hydrogel integrated with mesenchymal stem cell-derived endothelial cells to construct vascularized tissue in vivo. Regen Biomater 2021; 8:rbab051. [PMID: 34603743 PMCID: PMC8481010 DOI: 10.1093/rb/rbab051] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Revised: 08/08/2021] [Accepted: 08/30/2021] [Indexed: 12/12/2022] Open
Abstract
The ideal scaffold material of angiogenesis should have mechanical strength and provide appropriate physiological microporous structures to mimic the extracellular matrix environment. In this study, we constructed an integrated three-dimensional scaffold material using porous tantalum (pTa), gelatin nanoparticles (GNPs) hydrogel, and seeded with bone marrow mesenchymal stem cells (BMSCs)-derived endothelial cells (ECs) for vascular tissue engineering. The characteristics and biocompatibility of pTa and GNPs hydrogel were evaluated by mechanical testing, scanning electron microscopy, cell counting kit, and live-cell assay. The BMSCs-derived ECs were identified by flow cytometry and angiogenesis assay. BMSCs-derived ECs were seeded on the pTa-GNPs hydrogel scaffold and implanted subcutaneously in nude mice. Four weeks after the operation, the scaffold material was evaluated by histomorphology. The superior biocompatible ability of pTa-GNPs hydrogel scaffold was observed. Our in vivo results suggested that 28 days after implantation, the formation of the stable capillary-like network in scaffold material could be promoted significantly. The novel, integrated pTa-GNPs hydrogel scaffold is biocompatible with the host, and exhibits biomechanical and angiogenic properties. Moreover, combined with BMSCs-derived ECs, it could construct vascular engineered tissue in vivo. This study may provide a basis for applying pTa in bone regeneration and autologous BMSCs in tissue-engineered vascular grafts.
Collapse
Affiliation(s)
- Zhenhua Zhao
- Orthopaedic Department, Affiliated ZhongShan Hospital of Dalian University, No. 6 Jiefang Street, Zhongshan District, Dalian, Liaoning 116001, P. R. China
- National-Local Joint Engineering Laboratory for the Development of Orthopedic Implant Materials, Affiliated ZhongShan Hospital of Dalian University, No. 6 Jiefang Street, Zhongshan District, Dalian, Liaoning 116001, P. R. China
| | - Mang Wang
- Orthopaedic Department, Affiliated ZhongShan Hospital of Dalian University, No. 6 Jiefang Street, Zhongshan District, Dalian, Liaoning 116001, P. R. China
| | - Fei Shao
- Key State Laboratory of Fine Chemicals, School of Bioengineering, Dalian University of Technology, No. 2, Linggong Road, High-Tech District, Dalian 116024, P. R. China
| | - Ge Liu
- National-Local Joint Engineering Laboratory for the Development of Orthopedic Implant Materials, Affiliated ZhongShan Hospital of Dalian University, No. 6 Jiefang Street, Zhongshan District, Dalian, Liaoning 116001, P. R. China
- School of Mechanical Engineering, Dalian Jiaotong University, Dalian 116028, P. R. China
| | - Junlei Li
- National-Local Joint Engineering Laboratory for the Development of Orthopedic Implant Materials, Affiliated ZhongShan Hospital of Dalian University, No. 6 Jiefang Street, Zhongshan District, Dalian, Liaoning 116001, P. R. China
| | - Xiaowei Wei
- National-Local Joint Engineering Laboratory for the Development of Orthopedic Implant Materials, Affiliated ZhongShan Hospital of Dalian University, No. 6 Jiefang Street, Zhongshan District, Dalian, Liaoning 116001, P. R. China
| | - Xiuzhi Zhang
- National-Local Joint Engineering Laboratory for the Development of Orthopedic Implant Materials, Affiliated ZhongShan Hospital of Dalian University, No. 6 Jiefang Street, Zhongshan District, Dalian, Liaoning 116001, P. R. China
- Reproductive Medicine Centre, Affiliated ZhongShan Hospital of Dalian University, No. 6 Jiefang Street, Zhongshan District, Dalian, Liaoning 116001, P. R. China
| | - Jiahui Yang
- National-Local Joint Engineering Laboratory for the Development of Orthopedic Implant Materials, Affiliated ZhongShan Hospital of Dalian University, No. 6 Jiefang Street, Zhongshan District, Dalian, Liaoning 116001, P. R. China
| | - Fang Cao
- Department of Biomedical Engineering, Faculty of Electronic Information and Electronical Engineering, Dalian University of Technology, Dalian 116024, P. R. China
| | - Qiushi Wang
- Laboratory Animal Center, Affiliated ZhongShan Hospital of Dalian University, No. 6 Jiefang Street, Zhongshan District, Dalian, Liaoning 116001, P. R. China
| | - Huanan Wang
- Key State Laboratory of Fine Chemicals, School of Bioengineering, Dalian University of Technology, No. 2, Linggong Road, High-Tech District, Dalian 116024, P. R. China
| | - Dewei Zhao
- Orthopaedic Department, Affiliated ZhongShan Hospital of Dalian University, No. 6 Jiefang Street, Zhongshan District, Dalian, Liaoning 116001, P. R. China
- National-Local Joint Engineering Laboratory for the Development of Orthopedic Implant Materials, Affiliated ZhongShan Hospital of Dalian University, No. 6 Jiefang Street, Zhongshan District, Dalian, Liaoning 116001, P. R. China
| |
Collapse
|
20
|
Pirosa A, Tankus EB, Mainardi A, Occhetta P, Dönges L, Baum C, Rasponi M, Martin I, Barbero A. Modeling In Vitro Osteoarthritis Phenotypes in a Vascularized Bone Model Based on a Bone-Marrow Derived Mesenchymal Cell Line and Endothelial Cells. Int J Mol Sci 2021; 22:ijms22179581. [PMID: 34502489 PMCID: PMC8430538 DOI: 10.3390/ijms22179581] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 08/25/2021] [Accepted: 09/01/2021] [Indexed: 12/03/2022] Open
Abstract
The subchondral bone and its associated vasculature play an important role in the onset of osteoarthritis (OA). Integration of different aspects of the OA environment into multi-cellular and complex human, in vitro models is therefore needed to properly represent the pathology. In this study, we exploited a mesenchymal stromal cell line/endothelial cell co-culture to produce an in vitro human model of vascularized osteogenic tissue. A cocktail of inflammatory cytokines, or conditioned medium from mechanically-induced OA engineered microcartilage, was administered to this vascularized bone model to mimic the inflamed OA environment, hypothesizing that these treatments could induce the onset of specific pathological traits. Exposure to the inflammatory factors led to increased network formation by endothelial cells, reminiscent of the abnormal angiogenesis found in OA subchondral bone, demineralization of the constructs, and increased collagen production, signs of OA related bone sclerosis. Furthermore, inflammation led to augmented expression of osteogenic (alkaline phosphatase (ALP) and osteocalcin (OCN)) and angiogenic (vascular endothelial growth factor (VEGF)) genes. The treatment, with a conditioned medium from the mechanically-induced OA engineered microcartilage, also caused increased demineralization and expression of ALP, OCN, ADAMTS5, and VEGF; however, changes in network formation by endothelial cells were not observed in this second case, suggesting a possible different mechanism of action in inducing OA-like phenotypes. We propose that this vascularized bone model could represent a first step for the in vitro study of bone changes under OA mimicking conditions and possibly serve as a tool in testing anti-OA drugs.
Collapse
Affiliation(s)
- Alessandro Pirosa
- Department of Biomedicine, University Hospital Basel, University of Basel, 4056 Basel, Switzerland; (A.P.); (E.B.T.); (A.M.); (L.D.); (I.M.)
| | - Esma Bahar Tankus
- Department of Biomedicine, University Hospital Basel, University of Basel, 4056 Basel, Switzerland; (A.P.); (E.B.T.); (A.M.); (L.D.); (I.M.)
| | - Andrea Mainardi
- Department of Biomedicine, University Hospital Basel, University of Basel, 4056 Basel, Switzerland; (A.P.); (E.B.T.); (A.M.); (L.D.); (I.M.)
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, 20133 Milan, Italy; (P.O.); (M.R.)
- Department of Biomedical Engineering, University of Basel, 4123 Allschwil, Switzerland
| | - Paola Occhetta
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, 20133 Milan, Italy; (P.O.); (M.R.)
| | - Laura Dönges
- Department of Biomedicine, University Hospital Basel, University of Basel, 4056 Basel, Switzerland; (A.P.); (E.B.T.); (A.M.); (L.D.); (I.M.)
| | - Cornelia Baum
- Department of Orthopaedic Surgery and Traumatology, University Hospital Basel, 4031 Basel, Switzerland;
- Department of Research and Development, Schulthess Klinik Zurich, 8008 Zurich, Switzerland
| | - Marco Rasponi
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, 20133 Milan, Italy; (P.O.); (M.R.)
| | - Ivan Martin
- Department of Biomedicine, University Hospital Basel, University of Basel, 4056 Basel, Switzerland; (A.P.); (E.B.T.); (A.M.); (L.D.); (I.M.)
| | - Andrea Barbero
- Department of Biomedicine, University Hospital Basel, University of Basel, 4056 Basel, Switzerland; (A.P.); (E.B.T.); (A.M.); (L.D.); (I.M.)
- Correspondence:
| |
Collapse
|
21
|
Chirică IM, Enciu AM, Tite T, Dudău M, Albulescu L, Iconaru SL, Predoi D, Pasuk I, Enculescu M, Radu C, Mihalcea CG, Popa AC, Rusu N, Niţă S, Tănase C, Stan GE. The Physico-Chemical Properties and Exploratory Real-Time Cell Analysis of Hydroxyapatite Nanopowders Substituted with Ce, Mg, Sr, and Zn (0.5-5 at.%). MATERIALS 2021; 14:ma14143808. [PMID: 34300727 PMCID: PMC8305395 DOI: 10.3390/ma14143808] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 07/01/2021] [Accepted: 07/05/2021] [Indexed: 12/17/2022]
Abstract
Cation-substituted hydroxyapatite (HA), standalone or as a composite (blended with polymers or metals), is currently regarded as a noteworthy candidate material for bone repair/regeneration either in the form of powders, porous scaffolds or coatings for endo-osseous dental and orthopaedic implants. As a response to the numerous contradictions reported in literature, this work presents, in one study, the physico-chemical properties and the cytocompatibility response of single cation-doped (Ce, Mg, Sr or Zn) HA nanopowders in a wide concentration range (0.5–5 at.%). The modification of composition, morphology, and structure was multiparametrically monitored via energy dispersive X-ray, X-ray photoelectron, Fourier-transform infrared and micro-Raman spectroscopy methods, as well as by transmission electron microscopy and X-ray diffraction. From a compositional point of view, Ce and Sr were well-incorporated in HA, while slight and pronounced deviations were observed for Mg and Zn, respectively. The change of the lattice parameters, crystallite size, and substituting cation occupation factors either in the Ca(I) or Ca(II) sites were further determined. Sr produced the most important HA structural changes. The in vitro biological performance was evaluated by the (i) determination of leached therapeutic cations (by inductively coupled plasma mass spectrometry) and (ii) assessment of cell behaviour by both conventional assays (e.g., proliferation—3-(4,5-dimethyl thiazol-2-yl) 5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium assay; cytotoxicity—lactate dehydrogenase release assay) and, for the first time, real-time cell analysis (RTCA). Three cell lines were employed: fibroblast, osteoblast, and endothelial. When monophasic, the substituted HA supported the cells’ viability and proliferation without signs of toxicity. The RTCA results indicate the excellent adherence of cells. The study strived to offer a perspective on the behaviour of Ce-, Mg-, Sr-, or Zn-substituted HAs and to deliver a well-encompassing viewpoint on their effects. This can be highly important for the future development of such bioceramics, paving the road toward the identification of candidates with highly promising therapeutic effects.
Collapse
Affiliation(s)
- Iuliana Maria Chirică
- National Institute of Materials Physics, RO-077125 Măgurele, Romania; (I.M.C.); (S.L.I.); (D.P.); (I.P.); (M.E.); (C.R.); (C.G.M.); (A.-C.P.)
- Faculty of Physics, University of Bucharest, RO-077125 Măgurele, Romania
| | - Ana-Maria Enciu
- “Victor Babes” National Institute of Pathology, RO-050096 Bucharest, Romania; (A.-M.E.); (M.D.); (L.A.)
- Department of Cellular Biology and Histology, “Carol Davila” University of Medicine and Pharmacy, RO-050047 Bucharest, Romania
| | - Teddy Tite
- National Institute of Materials Physics, RO-077125 Măgurele, Romania; (I.M.C.); (S.L.I.); (D.P.); (I.P.); (M.E.); (C.R.); (C.G.M.); (A.-C.P.)
- Correspondence: (T.T.); (C.T.); (G.E.S.); Tel./Fax: +40-21-319-4528 (C.T.); Tel.: +40-21-241-8128 (G.E.S.); Fax: +40-21-369-0177 (G.E.S.)
| | - Maria Dudău
- “Victor Babes” National Institute of Pathology, RO-050096 Bucharest, Romania; (A.-M.E.); (M.D.); (L.A.)
- Department of Cellular Biology and Histology, “Carol Davila” University of Medicine and Pharmacy, RO-050047 Bucharest, Romania
| | - Lucian Albulescu
- “Victor Babes” National Institute of Pathology, RO-050096 Bucharest, Romania; (A.-M.E.); (M.D.); (L.A.)
| | - Simona Liliana Iconaru
- National Institute of Materials Physics, RO-077125 Măgurele, Romania; (I.M.C.); (S.L.I.); (D.P.); (I.P.); (M.E.); (C.R.); (C.G.M.); (A.-C.P.)
| | - Daniela Predoi
- National Institute of Materials Physics, RO-077125 Măgurele, Romania; (I.M.C.); (S.L.I.); (D.P.); (I.P.); (M.E.); (C.R.); (C.G.M.); (A.-C.P.)
| | - Iuliana Pasuk
- National Institute of Materials Physics, RO-077125 Măgurele, Romania; (I.M.C.); (S.L.I.); (D.P.); (I.P.); (M.E.); (C.R.); (C.G.M.); (A.-C.P.)
| | - Monica Enculescu
- National Institute of Materials Physics, RO-077125 Măgurele, Romania; (I.M.C.); (S.L.I.); (D.P.); (I.P.); (M.E.); (C.R.); (C.G.M.); (A.-C.P.)
| | - Cristian Radu
- National Institute of Materials Physics, RO-077125 Măgurele, Romania; (I.M.C.); (S.L.I.); (D.P.); (I.P.); (M.E.); (C.R.); (C.G.M.); (A.-C.P.)
- Faculty of Physics, University of Bucharest, RO-077125 Măgurele, Romania
| | - Cătălina Gabriela Mihalcea
- National Institute of Materials Physics, RO-077125 Măgurele, Romania; (I.M.C.); (S.L.I.); (D.P.); (I.P.); (M.E.); (C.R.); (C.G.M.); (A.-C.P.)
- Faculty of Physics, University of Bucharest, RO-077125 Măgurele, Romania
| | - Adrian-Claudiu Popa
- National Institute of Materials Physics, RO-077125 Măgurele, Romania; (I.M.C.); (S.L.I.); (D.P.); (I.P.); (M.E.); (C.R.); (C.G.M.); (A.-C.P.)
| | - Nicoleta Rusu
- National Institute for Chemical Pharmaceutical Research and Development, RO-031299 Bucharest, Romania; (N.R.); (S.N.)
| | - Sultana Niţă
- National Institute for Chemical Pharmaceutical Research and Development, RO-031299 Bucharest, Romania; (N.R.); (S.N.)
| | - Cristiana Tănase
- “Victor Babes” National Institute of Pathology, RO-050096 Bucharest, Romania; (A.-M.E.); (M.D.); (L.A.)
- “Nicolae Cajal” Institute, “Titu Maiorescu” University, RO-004051 Bucharest, Romania
- Correspondence: (T.T.); (C.T.); (G.E.S.); Tel./Fax: +40-21-319-4528 (C.T.); Tel.: +40-21-241-8128 (G.E.S.); Fax: +40-21-369-0177 (G.E.S.)
| | - George E. Stan
- National Institute of Materials Physics, RO-077125 Măgurele, Romania; (I.M.C.); (S.L.I.); (D.P.); (I.P.); (M.E.); (C.R.); (C.G.M.); (A.-C.P.)
- Correspondence: (T.T.); (C.T.); (G.E.S.); Tel./Fax: +40-21-319-4528 (C.T.); Tel.: +40-21-241-8128 (G.E.S.); Fax: +40-21-369-0177 (G.E.S.)
| |
Collapse
|
22
|
Zhang X, Jiang X, Jiang S, Cai X, Yu S, Pei G. Schwann cells promote prevascularization and osteogenesis of tissue-engineered bone via bone marrow mesenchymal stem cell-derived endothelial cells. Stem Cell Res Ther 2021; 12:382. [PMID: 34233721 PMCID: PMC8261922 DOI: 10.1186/s13287-021-02433-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 06/06/2021] [Indexed: 12/18/2022] Open
Abstract
Background Tissue-engineered bone grafts (TEBGs) that undergo vascularization and neurotization evolve into functioning bone tissue. Previously, we verified that implanting sensory nerve tracts into TEBGs promoted osteogenesis. However, the precise mechanisms and interaction between seed cells were not explored. In this study, we hypothesized that neurotization may influence the osteogenesis of TEBGs through vascularization. Methods We cultured rat Schwann cells (SCs), aortic endothelial cells (AECs), and bone marrow-derived mesenchymal stem cells (BM-MSCs) and then obtained BM-MSC-derived induced endothelial cells (IECs) and induced osteoblasts (IOBs). IECs and AECs were cultured in an SC-conditioned medium (SC-CM) to assess proliferation, migration, capillary-like tube formation, and angiogenesis, and the vascular endothelial growth factor (VEGF) levels in the supernatants were detected. We established an indirect coculture model to detect the expression of nestin and VEGF receptors in IECs and tissue inhibitor of metalloproteinase (TIMP)-2 in SCs. Then, SCs, IECs, and IOBs were labeled and loaded into a β-tricalcium phosphate scaffold to induce prevascularization, and the scaffold was implanted into a 6-mm-long defect of rat femurs. Three groups were set up according to the loaded cells: I, SCs, and IECs (coculture for 3 days) plus IOBs; II, IECs (culture for 3 days) plus IOBs; III, IOBs. Nestin and TIMP-2 expression and osteogenesis of TEBGs were evaluated at 12 weeks post-implantation through histological and radiological assessments. Results We found that SC-CM promoted IEC proliferation, migration, capillary-like tube formation, and angiogenesis, but no similar effects were observed for AECs. IECs expressed nestin extensively, while AECs barely expressed nestin, and SC-CM promoted the VEGF secretion of IECs. In the coculture model, SCs promoted nestin and VEGF receptor expression in IECs, and IECs inhibited TIMP-2 expression in SCs. The promotion of prevascularized TEBGs by SCs and IECs in group I augmented new bone formation at 6 and 12 weeks. Nestin expression was higher in group I than in the other groups, while TIMP-2 expression was lower at 12 weeks. Conclusions This study demonstrated that SCs can promote TEBG osteogenesis via IECs and further revealed the related specific characteristics of IECs, providing preliminary cytological evidence for neurotization of TEBGs. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-021-02433-3.
Collapse
Affiliation(s)
- Xinxin Zhang
- Department of Orthopaedics, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 17 Panjiayuannanli, Chaoyang District, Beijing, 100021, China
| | - Xiaorui Jiang
- Department of Hand and Foot Orthopaedics, Yantai Yuhuangding Hospital, Qingdao University Medical College, Yantai, Shandong, China
| | - Shan Jiang
- Department of Clinical Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Xiyu Cai
- Department of Orthopedics, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, Guangdong, China
| | - Shengji Yu
- Department of Orthopaedics, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 17 Panjiayuannanli, Chaoyang District, Beijing, 100021, China.
| | - Guoxian Pei
- Southern University of Science and Technology Hospital, No. 6019 Liuxian Street, Xili Avenue, Nanshan District, Shenzhen, 518055, Guangdong, China.
| |
Collapse
|
23
|
Whelan IT, Moeendarbary E, Hoey DA, Kelly DJ. Biofabrication of vasculature in microphysiological models of bone. Biofabrication 2021; 13. [PMID: 34034238 DOI: 10.1088/1758-5090/ac04f7] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 05/25/2021] [Indexed: 11/12/2022]
Abstract
Bone contains a dense network of blood vessels that are essential to its homoeostasis, endocrine function, mineral metabolism and regenerative functions. In addition, bone vasculature is implicated in a number of prominent skeletal diseases, and bone has high affinity for metastatic cancers. Despite vasculature being an integral part of bone physiology and pathophysiology, it is often ignored or oversimplified inin vitrobone models. However, 3D physiologically relevant vasculature can now be engineeredin vitro, with microphysiological systems (MPS) increasingly being used as platforms for engineering this physiologically relevant vasculature. In recent years, vascularised models of bone in MPSs systems have been reported in the literature, representing the beginning of a possible technological step change in how bone is modelledin vitro. Vascularised bone MPSs is a subfield of bone research in its nascency, however given the impact of MPSs has had inin vitroorgan modelling, and the crucial role of vasculature to bone physiology, these systems stand to have a substantial impact on bone research. However, engineering vasculature within the specific design restraints of the bone niche is significantly challenging given the different requirements for engineering bone and vasculature. With this in mind, this paper aims to serve as technical guidance for the biofabrication of vascularised bone tissue within MPS devices. We first discuss the key engineering and biological considerations for engineering more physiologically relevant vasculaturein vitrowithin the specific design constraints of the bone niche. We next explore emerging applications of vascularised bone MPSs, and conclude with a discussion on the current status of vascularised bone MPS biofabrication and suggest directions for development of next generation vascularised bone MPSs.
Collapse
|
24
|
Sevari SP, Ansari S, Moshaverinia A. A narrative overview of utilizing biomaterials to recapitulate the salient regenerative features of dental-derived mesenchymal stem cells. Int J Oral Sci 2021; 13:22. [PMID: 34193832 PMCID: PMC8245503 DOI: 10.1038/s41368-021-00126-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 05/26/2021] [Accepted: 06/03/2021] [Indexed: 02/06/2023] Open
Abstract
Tissue engineering approaches have emerged recently to circumvent many limitations associated with current clinical practices. This elegant approach utilizes a natural/synthetic biomaterial with optimized physiomechanical properties to serve as a vehicle for delivery of exogenous stem cells and bioactive factors or induce local recruitment of endogenous cells for in situ tissue regeneration. Inspired by the natural microenvironment, biomaterials could act as a biomimetic three-dimensional (3D) structure to help the cells establish their natural interactions. Such a strategy should not only employ a biocompatible biomaterial to induce new tissue formation but also benefit from an easily accessible and abundant source of stem cells with potent tissue regenerative potential. The human teeth and oral cavity harbor various populations of mesenchymal stem cells (MSCs) with self-renewing and multilineage differentiation capabilities. In the current review article, we seek to highlight recent progress and future opportunities in dental MSC-mediated therapeutic strategies for tissue regeneration using two possible approaches, cell transplantation and cell homing. Altogether, this paper develops a general picture of current innovative strategies to employ dental-derived MSCs combined with biomaterials and bioactive factors for regenerating the lost or defective tissues and offers information regarding the available scientific data and possible applications.
Collapse
Affiliation(s)
- Sevda Pouraghaei Sevari
- Weintraub Center for Reconstructive Biotechnology, Division of Advanced Prosthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, CA, USA
| | - Sahar Ansari
- Weintraub Center for Reconstructive Biotechnology, Division of Advanced Prosthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, CA, USA
| | - Alireza Moshaverinia
- Weintraub Center for Reconstructive Biotechnology, Division of Advanced Prosthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, CA, USA.
- California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
25
|
Bartolotti I, Roseti L, Petretta M, Grigolo B, Desando G. A Roadmap of In Vitro Models in Osteoarthritis: A Focus on Their Biological Relevance in Regenerative Medicine. J Clin Med 2021; 10:1920. [PMID: 33925222 PMCID: PMC8124812 DOI: 10.3390/jcm10091920] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 04/20/2021] [Accepted: 04/26/2021] [Indexed: 01/15/2023] Open
Abstract
Osteoarthritis (OA) is a multifaceted musculoskeletal disorder, with a high prevalence worldwide. Articular cartilage and synovial membrane are among the main biological targets in the OA microenvironment. Gaining more knowledge on the accuracy of preclinical in vitro OA models could open innovative avenues in regenerative medicine to bridge major gaps, especially in translation from animals to humans. Our methodological approach entailed searches on Scopus, the Web of Science Core Collection, and EMBASE databases to select the most relevant preclinical in vitro models for studying OA. Predicting the biological response of regenerative strategies requires developing relevant preclinical models able to mimic the OA milieu influencing tissue responses and organ complexity. In this light, standard 2D culture models lack critical properties beyond cell biology, while animal models suffer from several limitations due to species differences. In the literature, most of the in vitro models only recapitulate a tissue compartment, by providing fragmented results. Biotechnological advances may enable scientists to generate new in vitro models that combine easy manipulation and organ complexity. Here, we review the state-of-the-art of preclinical in vitro models in OA and outline how the different preclinical systems (inflammatory/biomechanical/microfluidic models) may be valid tools in regenerative medicine, describing their pros and cons. We then discuss the prospects of specific and combinatorial models to predict biological responses following regenerative approaches focusing on mesenchymal stromal cells (MSCs)-based therapies to reduce animal testing.
Collapse
Affiliation(s)
- Isabella Bartolotti
- Laboratorio RAMSES, IRCCS Istituto Ortopedico Rizzoli, via di Barbiano 1/10, 40136 Bologna, Italy; (I.B.); (M.P.); (B.G.)
| | - Livia Roseti
- Laboratorio RAMSES, IRCCS Istituto Ortopedico Rizzoli, via di Barbiano 1/10, 40136 Bologna, Italy; (I.B.); (M.P.); (B.G.)
| | - Mauro Petretta
- Laboratorio RAMSES, IRCCS Istituto Ortopedico Rizzoli, via di Barbiano 1/10, 40136 Bologna, Italy; (I.B.); (M.P.); (B.G.)
- RegenHu Company, Z.I Du Vivier 22, 1690 Villaz-St-Pierre, Switzerland
| | - Brunella Grigolo
- Laboratorio RAMSES, IRCCS Istituto Ortopedico Rizzoli, via di Barbiano 1/10, 40136 Bologna, Italy; (I.B.); (M.P.); (B.G.)
| | - Giovanna Desando
- Laboratorio RAMSES, IRCCS Istituto Ortopedico Rizzoli, via di Barbiano 1/10, 40136 Bologna, Italy; (I.B.); (M.P.); (B.G.)
| |
Collapse
|
26
|
Pirosa A, Gottardi R, Alexander PG, Puppi D, Chiellini F, Tuan RS. An in vitro chondro-osteo-vascular triphasic model of the osteochondral complex. Biomaterials 2021; 272:120773. [PMID: 33798958 DOI: 10.1016/j.biomaterials.2021.120773] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 03/15/2021] [Accepted: 03/19/2021] [Indexed: 01/06/2023]
Abstract
The generation of engineered models of the osteochondral complex to study its pathologies and develop possible treatments is hindered by the distinctly different properties of articular cartilage and subchondral bone, with the latter characterized by vascularization. In vitro models of the osteochondral complex have been mainly engineered as biphasic constructs containing just cartilage and bone cells, a condition very dissimilar from the in vivo environment. The different cellular components of the osteochondral complex are governed by interacting biochemical signaling; hence, to study the crosstalk among chondrocytes, osteoblasts, and endothelial cells, we have developed a novel triphasic model of the osteochondral tissue interface. Wet-spun poly(ε-caprolactone) (PCL) and PCL/hydroxyapatite (HA) scaffolds in combination with a methacrylated gelatin (gelMA) hydrogel were used as the polymeric backbone of the constructs. The scaffold components were engineered with human bone marrow derived mesenchymal stem cells (hMSCs) and human umbilical vein endothelial cells (HUVECs), and differentiated using a dual chamber microphysiological system (MPS) bioreactor that allows the simultaneous, separate flow of media of different compositions for induced differentiation of each compartment towards a cartilaginous or osseous lineage. Within the engineered Microphysiological Vascularized Osteochondral System, hMSCs showed spatially distinct chondrogenic and osteogenic markers in terms of histology and gene expression. HUVECs formed a stable capillary-like network in the engineered bone compartment and enhanced both chondrogenic and osteogenic differentiation of hMSCs, resulting in the generation of an in vitro system that mimics a vascularized osteochondral interface tissue.
Collapse
Affiliation(s)
- Alessandro Pirosa
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; BIOlab Research Group, Department of Chemistry and Industrial Chemistry, University of Pisa, Pisa, Italy
| | - Riccardo Gottardi
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Ri.MED Foundation, Palermo, Italy
| | - Peter G Alexander
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Dario Puppi
- BIOlab Research Group, Department of Chemistry and Industrial Chemistry, University of Pisa, Pisa, Italy
| | - Federica Chiellini
- BIOlab Research Group, Department of Chemistry and Industrial Chemistry, University of Pisa, Pisa, Italy
| | - Rocky S Tuan
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| |
Collapse
|
27
|
Amler AK, Thomas A, Tüzüner S, Lam T, Geiger MA, Kreuder AE, Palmer C, Nahles S, Lauster R, Kloke L. 3D bioprinting of tissue-specific osteoblasts and endothelial cells to model the human jawbone. Sci Rep 2021; 11:4876. [PMID: 33649412 PMCID: PMC7921109 DOI: 10.1038/s41598-021-84483-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 02/15/2021] [Indexed: 02/06/2023] Open
Abstract
Jawbone differs from other bones in many aspects, including its developmental origin and the occurrence of jawbone-specific diseases like MRONJ (medication-related osteonecrosis of the jaw). Although there is a strong need, adequate in vitro models of this unique environment are sparse to date. While previous approaches are reliant e.g. on scaffolds or spheroid culture, 3D bioprinting enables free-form fabrication of complex living tissue structures. In the present work, production of human jawbone models was realised via projection-based stereolithography. Constructs were bioprinted containing primary jawbone-derived osteoblasts and vasculature-like channel structures optionally harbouring primary endothelial cells. After 28 days of cultivation in growth medium or osteogenic medium, expression of cell type-specific markers was confirmed on both the RNA and protein level, while prints maintained their overall structure. Survival of endothelial cells in the printed channels, co-cultured with osteoblasts in medium without supplementation of endothelial growth factors, was demonstrated. Constructs showed not only mineralisation, being one of the characteristics of osteoblasts, but also hinted at differentiation to an osteocyte phenotype. These results indicate the successful biofabrication of an in vitro model of the human jawbone, which presents key features of this special bone entity and hence appears promising for application in jawbone-specific research.
Collapse
Affiliation(s)
- Anna-Klara Amler
- Cellbricks GmbH, Gustav-Meyer-Allee 25, 13355, Berlin, Germany. .,Department of Medical Biotechnology, Technische Universität Berlin, Gustav-Meyer-Allee 25, 13355, Berlin, Germany.
| | - Alexander Thomas
- Cellbricks GmbH, Gustav-Meyer-Allee 25, 13355, Berlin, Germany.,Department of Medical Biotechnology, Technische Universität Berlin, Gustav-Meyer-Allee 25, 13355, Berlin, Germany
| | - Selin Tüzüner
- Cellbricks GmbH, Gustav-Meyer-Allee 25, 13355, Berlin, Germany.,Department of Medical Biotechnology, Technische Universität Berlin, Gustav-Meyer-Allee 25, 13355, Berlin, Germany
| | - Tobias Lam
- Cellbricks GmbH, Gustav-Meyer-Allee 25, 13355, Berlin, Germany
| | | | - Anna-Elisabeth Kreuder
- Cellbricks GmbH, Gustav-Meyer-Allee 25, 13355, Berlin, Germany.,Department of Medical Biotechnology, Technische Universität Berlin, Gustav-Meyer-Allee 25, 13355, Berlin, Germany
| | - Chris Palmer
- Cellbricks GmbH, Gustav-Meyer-Allee 25, 13355, Berlin, Germany
| | - Susanne Nahles
- Department of Oral- and Maxillofacial Surgery, Charité Campus Virchow, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Roland Lauster
- Department of Medical Biotechnology, Technische Universität Berlin, Gustav-Meyer-Allee 25, 13355, Berlin, Germany
| | - Lutz Kloke
- Cellbricks GmbH, Gustav-Meyer-Allee 25, 13355, Berlin, Germany
| |
Collapse
|
28
|
Veronesi F, Torricelli P, Martini L, Tschon M, Giavaresi G, Bellini D, Casagranda V, Alemani F, Fini M. An alternative ex vivo method to evaluate the osseointegration of Ti-6Al-4V alloy also combined with collagen. Biomed Mater 2021; 16:025007. [PMID: 33445161 DOI: 10.1088/1748-605x/abdbda] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Due to the increasing number of orthopedic implantation surgery and advancements in biomaterial manufacturing, chemistry and topography, there is an increasing need of reliable and rapid methods for the preclinical investigation of osseointegration and bone ingrowth. Implant surface composition and topography increase osteogenicity, osteoinductivity, osteoconductivity and osseointegration of a prosthesis. Among the biomaterials used to manufacture an orthopedic prosthesis, titanium alloy (Ti-6Al-4V) is the most used. Type I collagen (COLL I) induces cell function, adhesion, differentiation and bone extracellular matrix component secretion and it is reported to improve osseointegration if immobilized on the alloy surface. The aim of the present study was to evaluate the feasibility of an alternative ex vivo model, developed by culturing rabbit cortical bone segments with Ti-6Al-4V alloy cylinders (Ti-POR), fabricated through the process of electron beam melting (EBM), to evaluate osseointegration. In addition, a comparison was made with Ti-POR coated with COLL I (Ti-POR-COLL) to evaluate osseointegration in terms of bone-to-implant contact (BIC) and new bone formation (nBAr/TAr) at 30, 60 and 90 d of culture. After 30 and 60 d of culture, BIC and nBAr/TAr resulted significantly higher in Ti-POR-COLL implants than in Ti-POR. No differences have been found at 90 d of culture. With the developed model it was possible to distinguish the biomaterial properties and behavior. This study defined and confirmed for the first time the validity of the alternative ex vivo method to evaluate osseointegration and that COLL I improves osseointegration and bone growth of Ti-6Al-4V fabricated through EBM.
Collapse
Affiliation(s)
- Francesca Veronesi
- Complex Structure of Surgical Sciences and Technologies, IRCCS Istituto Ortopedico Rizzoli, Via Di Barbiano 1/10, 40136 Bologna, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Yang G, Mahadik B, Choi JY, Yu JR, Mollot T, Jiang B, He X, Fisher JP. Fabrication of centimeter-sized 3D constructs with patterned endothelial cells through assembly of cell-laden microbeads as a potential bone graft. Acta Biomater 2021; 121:204-213. [PMID: 33271356 DOI: 10.1016/j.actbio.2020.11.040] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 11/24/2020] [Accepted: 11/25/2020] [Indexed: 02/07/2023]
Abstract
Modular tissue engineering is a promising biofabrication strategy to create engineered bone grafts in a bottom-up manner, in which cell-laden micro-modules are prepared as basic building blocks to assemble macroscopic tissues via different integrating mechanisms. In this study, we prepared collagen microbeads loaded with human bone marrow derived mesenchymal stem cells (BMSCs) using a microfluidic approach. The cell-laden microbeads were characterized for size change, cell activity, osteogenesis, as well as their self-assembly properties to generate centimeter-sized constructs. Moreover, using the cell-laden beads as a supporting medium, induced pluripotent stem cell-derived endothelial cells (iPSC-EC) were patterned inside bead aggregates through extrusion-based 3D printing. This fabrication approach that combines modular tissue engineering and supports 3D printing has the potential to create 3D engineered bone grafts with a pre-existing, customized vasculature.
Collapse
Affiliation(s)
- Guang Yang
- Tissue Engineering & Biomaterials Laboratory, Fischell Department of Bioengineering, A. James Clark School of Engineering, University of Maryland, College Park, MD 20742, United States; NIBIB/NIH Center for Engineering Complex Tissues, University of Maryland, College Park, MD 20742, United States
| | - Bhushan Mahadik
- Tissue Engineering & Biomaterials Laboratory, Fischell Department of Bioengineering, A. James Clark School of Engineering, University of Maryland, College Park, MD 20742, United States; NIBIB/NIH Center for Engineering Complex Tissues, University of Maryland, College Park, MD 20742, United States
| | - Ji Young Choi
- Tissue Engineering & Biomaterials Laboratory, Fischell Department of Bioengineering, A. James Clark School of Engineering, University of Maryland, College Park, MD 20742, United States; NIBIB/NIH Center for Engineering Complex Tissues, University of Maryland, College Park, MD 20742, United States
| | - Justine R Yu
- Tissue Engineering & Biomaterials Laboratory, Fischell Department of Bioengineering, A. James Clark School of Engineering, University of Maryland, College Park, MD 20742, United States; NIBIB/NIH Center for Engineering Complex Tissues, University of Maryland, College Park, MD 20742, United States; University of Maryland School of Medicine, Baltimore, MD 21201, United States
| | - Trevor Mollot
- Tissue Engineering & Biomaterials Laboratory, Fischell Department of Bioengineering, A. James Clark School of Engineering, University of Maryland, College Park, MD 20742, United States
| | - Bin Jiang
- Fischell Department of Bioengineering, A. James Clark School of Engineering, University of Maryland, College Park, MD 20742, United States
| | - Xiaoming He
- Fischell Department of Bioengineering, A. James Clark School of Engineering, University of Maryland, College Park, MD 20742, United States
| | - John P Fisher
- Tissue Engineering & Biomaterials Laboratory, Fischell Department of Bioengineering, A. James Clark School of Engineering, University of Maryland, College Park, MD 20742, United States; NIBIB/NIH Center for Engineering Complex Tissues, University of Maryland, College Park, MD 20742, United States.
| |
Collapse
|
30
|
Hashemi S, Mohammadi Amirabad L, Farzad-Mohajeri S, Rezai Rad M, Fahimipour F, Ardeshirylajimi A, Dashtimoghadam E, Salehi M, Soleimani M, Dehghan MM, Tayebi L, Khojasteh A. Comparison of osteogenic differentiation potential of induced pluripotent stem cells and buccal fat pad stem cells on 3D-printed HA/β-TCP collagen-coated scaffolds. Cell Tissue Res 2021; 384:403-421. [PMID: 33433691 DOI: 10.1007/s00441-020-03374-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Accepted: 12/02/2020] [Indexed: 01/18/2023]
Abstract
Production of a 3D bone construct with high-yield differentiated cells using an appropriate cell source provides a reliable strategy for different purposes such as therapeutic screening of the drugs. Although adult stem cells can be a good source, their application is limited due to invasive procedure of their isolation and low yield of differentiation. Patient-specific human-induced pluripotent stem cells (hiPSCs) can be an alternative due to their long-term self-renewal capacity and pluripotency after several passages, resolving the requirement of a large number of progenitor cells. In this study, a new biphasic 3D-printed collagen-coated HA/β-TCP scaffold was fabricated to provide a 3D environment for the cells. The fabricated scaffolds were characterized by the 3D laser scanning digital microscopy, X-ray diffraction, Fourier transform infrared spectroscopy, and mechanical test. Then, the osteogenesis potential of the hiPSC-seeded scaffolds was investigated compared to the buccal fat pad stem cell (BFPSC)-seeded scaffolds through in vitro and in vivo studies. In vitro results demonstrated up-regulated expressions of osteogenesis-related genes of RUNX2, ALP, BMP2, and COL1 compared to the BFPSC-seeded scaffolds. In vivo results on calvarial defects in the rats confirmed a higher bone formation in the hiPSC-seeded scaffolds compared to the BFPSC-seeded groups. The immunofluorescence assay also showed higher expression levels of collagen I and osteocalcin proteins in the hiPSC-seeded scaffolds. It can be concluded that using the hiPSC-seeded scaffolds can lead to a high yield of osteogenesis, and the hiPSCs can be used as a superior stem cell source compared to BFPSCs for bone-like construct bioengineering.
Collapse
Affiliation(s)
- Sheida Hashemi
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Saeed Farzad-Mohajeri
- Department of Surgery and Radiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran.,Institute of Biomedical Research, University of Tehran, Tehran, Iran
| | - Maryam Rezai Rad
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Abdolreza Ardeshirylajimi
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Mohammad Salehi
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Masoud Soleimani
- Department of Hematology, Faculty of Medical Science, Tarbiat Modares University, Tehran, Iran
| | - Mohammad Mehdi Dehghan
- Department of Surgery and Radiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran.,Institute of Biomedical Research, University of Tehran, Tehran, Iran
| | - Lobat Tayebi
- Marquette University School of Dentistry, Milwaukee, WI, 53233, USA
| | - Arash Khojasteh
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
31
|
Zimmerling A, Chen X. Bioprinting for combating infectious diseases. BIOPRINTING (AMSTERDAM, NETHERLANDS) 2020; 20:e00104. [PMID: 33015403 PMCID: PMC7521216 DOI: 10.1016/j.bprint.2020.e00104] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 09/15/2020] [Accepted: 09/24/2020] [Indexed: 12/18/2022]
Abstract
Infectious diseases have the ability to impact health on a global scale, as is being demonstrated by the current coronavirus disease 2019 (COVID-19) pandemic. The strenuous circumstances related to this global health crisis have been highlighting the challenges faced by the biomedical field in combating infectious diseases. Notably, printing technologies have advanced rapidly over the last decades, allowing for the incorporation of living cells in the printing process (or bioprinting) to create constructs that are able to serve as in vitro tissue or virus-disease models in combating infectious diseases. This paper describes applications of bioprinting in addressing the challenges faced in combating infectious diseases, with a specific focus on in vitro modelling and on development of therapeutic agents and vaccines. Integration of these technologies may allow for a more efficient and effective response to current and future pandemics.
Collapse
Affiliation(s)
- Amanda Zimmerling
- Division of Biomedical Engineering, College of Engineering, University of Saskatchewan, Saskatoon, SK, Canada
| | - Xiongbiao Chen
- Division of Biomedical Engineering, College of Engineering, University of Saskatchewan, Saskatoon, SK, Canada
- Department of Mechanical Engineering, College of Engineering, University of Saskatchewan, Saskatoon, SK, Canada
| |
Collapse
|
32
|
Zidarič T, Milojević M, Vajda J, Vihar B, Maver U. Cultured Meat: Meat Industry Hand in Hand with Biomedical Production Methods. FOOD ENGINEERING REVIEWS 2020. [DOI: 10.1007/s12393-020-09253-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
33
|
Amelia F, Abbas B, Darwis D, Estuningsih S, Noviana D. Effects of bone types, particle sizes, and gamma irradiation doses in feline demineralized freeze-dried bone allograft. Vet World 2020; 13:1536-1543. [PMID: 33061224 PMCID: PMC7522947 DOI: 10.14202/vetworld.2020.1536-1543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Accepted: 06/09/2020] [Indexed: 11/16/2022] Open
Abstract
Background and Aim: Fracture cases significantly increase recently, demanding high quality of bone graft materials. This research aimed to evaluate the effects of bone types, particle sizes, and gamma irradiation doses on morphological performance and cell viability of feline demineralized freeze-dried bone allograft (DFDBA) through an in vitro study. Materials and Methods: Feline DFDBA derived from feline cortical and cancellous long bones was processed into four different sizes: Group A (larger than 1000 µm), B (841-1000 µm), C (420-840 µm), and D (250-419 µm) for each type of bones. The materials were then irradiated with two doses of gamma rays, 15 and 25 kGy, resulting in 16 variants of feline DFDBA. The surfaces of each material were then observed with the scanning electron microscope (SEM). The in vitro evaluation of feline DFDBA was then performed using 3-(4,5-dimethythiazol-2)-2,5-diphenyltetrazolium bromide (MTT) assay with calf pulmonary artery endothelial cells. Results: The MTT assay results showed that the lowest inhibition rate (14.67±9.17 %) achieved by feline DFDBA in Group A derived from cortical bones irradiated with 15 kGy. Group D generally showed high inhibition rate in both cancellous and cortical bones, irradiated with either 15 or 25 kGy. The SEM results showed that cancellous and cortical bones have numerous macropores and micropores structure in 170× and 3000×, respectively. Conclusion: The material derived from cortical bones in Group A (larger than 1000 µm in particle size) irradiated with 15 kGy is the best candidate for further development due to its abundance of micropores structure and ability in preserving the living cells.
Collapse
Affiliation(s)
- Frizky Amelia
- Program Study of Animal Biomedical Science, Graduate School of IPB University, Bogor, Jawa Barat 16680, Indonesia.,Diagnostic Imaging Center, Veterinary Teaching Hospital, Faculty of Veterinary Medicine IPB University, Bogor, Jawa Barat 16680, Indonesia
| | - Basril Abbas
- Centre for Isotopes and Radiation Application, National Nuclear Energy Agency (BATAN), Jakarta Selatan, DKI Jakarta 12440, Indonesia
| | - Darmawan Darwis
- Centre for Isotopes and Radiation Application, National Nuclear Energy Agency (BATAN), Jakarta Selatan, DKI Jakarta 12440, Indonesia
| | - Sri Estuningsih
- Department of Clinic Reproduction and Pathology, Faculty of Veterinary Medicine IPB University, Bogor, Jawa Barat 16680, Indonesia
| | - Deni Noviana
- Diagnostic Imaging Center, Veterinary Teaching Hospital, Faculty of Veterinary Medicine IPB University, Bogor, Jawa Barat 16680, Indonesia.,Department of Clinic Reproduction and Pathology, Faculty of Veterinary Medicine IPB University, Bogor, Jawa Barat 16680, Indonesia
| |
Collapse
|
34
|
Breathwaite E, Weaver J, Odanga J, dela Pena-Ponce M, Lee JB. 3D Bioprinted Osteogenic Tissue Models for In Vitro Drug Screening. Molecules 2020; 25:E3442. [PMID: 32751124 PMCID: PMC7435717 DOI: 10.3390/molecules25153442] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 07/26/2020] [Accepted: 07/27/2020] [Indexed: 02/08/2023] Open
Abstract
Metabolic bone disease affects hundreds of millions of people worldwide, and as a result, in vitro models of bone tissue have become essential tools to help analyze bone pathogenesis, develop drug screening, and test potential therapeutic strategies. Drugs that either promote or impair bone formation are in high demand for the treatment of metabolic bone diseases. These drugs work by targeting numerous signaling pathways responsible for regulating osteogenesis such as Hedgehog, Wnt/β-catenin, and PI3K-AKT. In this study, differentiated bone marrow-derived mesenchymal stem cell (BM-MSC) scaffold-free 3D bioprinted constructs and 2D monolayer cultures were utilized to screen four drugs predicted to either promote (Icariin and Purmorphamine) or impair osteogenesis (PD98059 and U0126). Osteogenic differentiation capacity was analyzed over a four week culture period by evaluating mineralization, alkaline phosphatase (ALP) activity, and osteogenesis related gene expression. Responses to drug treatment were observed in both 3D differentiated constructs and 2D monolayer cultures. After four weeks in culture, 3D differentiated constructs and 2D monolayer cultures treated with Icariin or Purmorphamine showed increased mineralization, ALP activity, and the gene expression of bone formation markers (BGLAP, SSP1, and COL1A1), signaling molecules (MAPK1, WNT1, and AKT1), and transcription factors (RUNX2 and GLI1) that regulate osteogenic differentiation relative to untreated. 3D differentiated constructs and 2D monolayer cultures treated with PD98059 or U0126 showed decreased mineralization, ALP activity, and the expression of the aforementioned genes BGLAP, SPP1, COL1A1, MAPK1, AKT1, RUNX2, and GLI1 relative to untreated. Differences in ALP activity and osteogenesis related gene expression relative to untreated cells cultured in a 2D monolayer were greater in 3D constructs compared to 2D monolayer cultures. These findings suggest that our bioprinted bone model system offers a more sensitive, biologically relevant drug screening platform than traditional 2D monolayer in vitro testing platforms.
Collapse
Affiliation(s)
| | | | | | | | - Jung Bok Lee
- Institute of Regenerative Medicine, LifeNet Health, 1864 Concert Drive, Virginia Beach, VA 23453, USA; (E.B.); (J.W.); (J.O.); (M.d.P.-P.)
| |
Collapse
|
35
|
Grémare A, Aussel A, Bareille R, Paiva Dos Santos B, Amédée J, Thébaud NB, Le Nihouannen D. A Unique Triculture Model to Study Osteoblasts, Osteoclasts, and Endothelial Cells. Tissue Eng Part C Methods 2020; 25:421-432. [PMID: 31169074 DOI: 10.1089/ten.tec.2018.0301] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
IMPACT STATEMENT In this article, we first developed a new medium to culture together primary human osteoblastic, osteoclastic, and endothelial cells (ECs) chosen to represent the three major bone cell tissues. Indeed, no study has been conducted on primary human cells and on the phenotype/activity retention of these three primary human cell types. Thus, we established an original triculture model with osteoblastic, osteoclastic, and ECs, where not only both cell phenotype and cell activity were maintained but also cell culture homeostasis. These promising results will permit further investigations to create in vitro conditions to mimic the bone microenvironment and analyze cell interactions in ex vivo studies.
Collapse
Affiliation(s)
- Agathe Grémare
- 1Université de Bordeaux, INSERM, Tissue Bioengineering, U1026, CHU Bordeaux, Services d'Odontologie et de Santé Buccale, F-33076, Bordeaux, France
| | - Audrey Aussel
- 1Université de Bordeaux, INSERM, Tissue Bioengineering, U1026, CHU Bordeaux, Services d'Odontologie et de Santé Buccale, F-33076, Bordeaux, France
| | - Reine Bareille
- 2Université de Bordeaux, INSERM, Tissue Bioengineering, U1026, F-33076, Bordeaux, France
| | - Bruno Paiva Dos Santos
- 2Université de Bordeaux, INSERM, Tissue Bioengineering, U1026, F-33076, Bordeaux, France
| | - Joelle Amédée
- 2Université de Bordeaux, INSERM, Tissue Bioengineering, U1026, F-33076, Bordeaux, France
| | - Noélie B Thébaud
- 1Université de Bordeaux, INSERM, Tissue Bioengineering, U1026, CHU Bordeaux, Services d'Odontologie et de Santé Buccale, F-33076, Bordeaux, France
| | - Damien Le Nihouannen
- 2Université de Bordeaux, INSERM, Tissue Bioengineering, U1026, F-33076, Bordeaux, France
| |
Collapse
|
36
|
Borciani G, Montalbano G, Baldini N, Cerqueni G, Vitale-Brovarone C, Ciapetti G. Co-culture systems of osteoblasts and osteoclasts: Simulating in vitro bone remodeling in regenerative approaches. Acta Biomater 2020; 108:22-45. [PMID: 32251782 DOI: 10.1016/j.actbio.2020.03.043] [Citation(s) in RCA: 107] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Revised: 03/20/2020] [Accepted: 03/30/2020] [Indexed: 02/08/2023]
Abstract
Bone is an extremely dynamic tissue, undergoing continuous remodeling for its whole lifetime, but its regeneration or augmentation due to bone loss or defects are not always easy to obtain. Bone tissue engineering (BTE) is a promising approach, and its success often relies on a "smart" scaffold, as a support to host and guide bone formation through bone cell precursors. Bone homeostasis is maintained by osteoblasts (OBs) and osteoclasts (OCs) within the basic multicellular unit, in a consecutive cycle of resorption and formation. Therefore, a functional scaffold should allow the best possible OB/OC cooperation for bone remodeling, as happens within the bone extracellular matrix in the body. In the present work OB/OC co-culture models, with and without scaffolds, are reviewed. These experimental systems are intended for different targets, including bone remodeling simulation, drug testing and the assessment of biomaterials and 3D scaffolds for BTE. As a consequence, several parameters, such as cell type, cell ratio, culture medium and inducers, culture times and setpoints, assay methods, etc. vary greatly. This review identifies and systematically reports the in vitro methods explored up to now, which, as they allow cellular communication, more closely resemble bone remodeling and/or the regeneration process in the framework of BTE. STATEMENT OF SIGNIFICANCE: Bone is a dynamic tissue under continuous remodeling, but spontaneous healing may fail in the case of excessive bone loss which often requires valid alternatives to conventional treatments to restore bone integrity, like bone tissue engineering (BTE). Pre-clinical evaluation of scaffolds for BTE requires in vitro testing where co-cultures combining innovative materials with osteoblasts (OBs) and osteoclasts (OCs) closely mimic the in vivo repair process. This review considers the direct and indirect OB/OC co-cultures relevant to BTE, from the early mouse-cell models to the recent bone regenerative systems. The co-culture modeling of bone microenvironment provides reliable information on bone cell cross-talk. Starting from improved knowledge on bone remodeling, bone disease mechanisms may be understood and new BTE solutions are designed.
Collapse
|
37
|
Nasello G, Alamán-Díez P, Schiavi J, Pérez MÁ, McNamara L, García-Aznar JM. Primary Human Osteoblasts Cultured in a 3D Microenvironment Create a Unique Representative Model of Their Differentiation Into Osteocytes. Front Bioeng Biotechnol 2020; 8:336. [PMID: 32391343 PMCID: PMC7193048 DOI: 10.3389/fbioe.2020.00336] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Accepted: 03/26/2020] [Indexed: 01/12/2023] Open
Abstract
Microengineered systems provide an in vitro strategy to explore the variability of individual patient response to tissue engineering products, since they prefer the use of primary cell sources representing the phenotype variability. Traditional in vitro systems already showed that primary human osteoblasts embedded in a 3D fibrous collagen matrix differentiate into osteocytes under specific conditions. Here, we hypothesized that translating this environment to the organ-on-a-chip scale creates a minimal functional unit to recapitulate osteoblast maturation toward osteocytes and matrix mineralization. Primary human osteoblasts were seeded in a type I collagen hydrogel, to establish the role of lower (2.5 × 105 cells/ml) and higher (1 × 106 cells/ml) cell density on their differentiation into osteocytes. A custom semi-automatic image analysis software was used to extract quantitative data on cellular morphology from brightfield images. The results are showing that cells cultured at a high density increase dendrite length over time, stop proliferating, exhibit dendritic morphology, upregulate alkaline phosphatase (ALP) activity, and express the osteocyte marker dental matrix protein 1 (DMP1). On the contrary, cells cultured at lower density proliferate over time, do not upregulate ALP and express the osteoblast marker bone sialoprotein 2 (BSP2) at all timepoints. Our work reveals that microengineered systems create unique conditions to capture the major aspects of osteoblast differentiation into osteocytes with a limited number of cells. We propose that the microengineered approach is a functional strategy to create a patient-specific bone tissue model and investigate the individual osteogenic potential of the patient bone cells.
Collapse
Affiliation(s)
- Gabriele Nasello
- Multiscale in Mechanical and Biological Engineering (M2BE), University of Zaragoza, Zaragoza, Spain.,Biomechanics Section, Department of Mechanical Engineering, KU Leuven, Leuven, Belgium
| | - Pilar Alamán-Díez
- Multiscale in Mechanical and Biological Engineering (M2BE), University of Zaragoza, Zaragoza, Spain
| | - Jessica Schiavi
- Mechanobiology and Medical Device Research Group (MMDRG), National University of Ireland Galway, Galway, Ireland
| | - María Ángeles Pérez
- Multiscale in Mechanical and Biological Engineering (M2BE), University of Zaragoza, Zaragoza, Spain
| | - Laoise McNamara
- Mechanobiology and Medical Device Research Group (MMDRG), National University of Ireland Galway, Galway, Ireland
| | - José Manuel García-Aznar
- Multiscale in Mechanical and Biological Engineering (M2BE), University of Zaragoza, Zaragoza, Spain
| |
Collapse
|
38
|
Shahabipour F, Oskuee RK, Dehghani H, Shokrgozar MA, Aninwene GE, Bonakdar S. Cell-cell interaction in a coculture system consisting of CRISPR/Cas9 mediated GFP knock-in HUVECs and MG-63 cells in alginate-GelMA based nanocomposites hydrogel as a 3D scaffold. J Biomed Mater Res A 2020; 108:1596-1606. [PMID: 32180319 DOI: 10.1002/jbm.a.36928] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2019] [Revised: 03/05/2020] [Accepted: 03/09/2020] [Indexed: 12/12/2022]
Abstract
The interaction between osteogenic and angiogenic cells through a coculturing system in biocompatible materials has been considered for successfully engineering vascularized bone tissue equivalents. In this study, we developed a hydrogel-blended scaffold consisted of gelatin methacryloyl (GelMA) and alginate enriched with hydroxyapatite nanoparticles (HAP) to model an in vitro prevascularized bone construct. The hydrogel-based scaffold revealed a higher mechanical stiffness than those of pure (GelMA), alginate, and (GelMA+ HAP) hydrogels. In the present study, we generated a green fluorescent protein (GFP) knock-in umbilical vein endothelial cells (HUVECs) cell line using the CRISPR/Cas9 technology. The GFP was inserted into the human-like ROSA locus of HUVECs genome. HUVECs expressing GFP were cocultured with OB-like cells (MG-63) within three-dimensionally (3D) fabricated hydrogel to investigate the response of cocultured osteoblasts and endothelial cells in a 3D structure. Cell viability under the 3D cocultured gel was higher than the 3D monocultured. Compared to the 3D monocultured condition, the cells were aligned and developed into the vessel-like structures. During 14 days of culture periods, the cells displayed actin protrusions by the formation of spike-like filopodia in the 3D cocultured model. Angiogenic and osteogenic-related genes such as CD31, vWF, and osteocalcin showed higher expression in the cocultured versus the monocultured. These results have collectively indicated that the 3D cocultured hydrogel facilitates interaction among cells, thereby having a greater effect on angiogenic and osteogenic properties in the absence of induction media.
Collapse
Affiliation(s)
| | - Reza K Oskuee
- Targeted Drug Delivery Research Center, Institute of Pharmaceutical Technology, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hesam Dehghani
- Division of Biotechnology, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, Mashhad, Iran.,Department of Basic Science, Faculty of Veterinary medicine, Ferdowsi University of Mashhad, Mashhad, Iran
| | | | - George E Aninwene
- Department of Bioengineering, University of California-Los Angeles, Los Angeles, California, USA.,Center for Minimally Invasive Therapeutics (C-MIT), University of California-Los Angeles, Los Angeles, California, USA
| | - Shahin Bonakdar
- National Cell Bank of Iran, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
39
|
Gardin C, Bosco G, Ferroni L, Quartesan S, Rizzato A, Tatullo M, Zavan B. Hyperbaric Oxygen Therapy Improves the Osteogenic and Vasculogenic Properties of Mesenchymal Stem Cells in the Presence of Inflammation In Vitro. Int J Mol Sci 2020; 21:ijms21041452. [PMID: 32093391 PMCID: PMC7073059 DOI: 10.3390/ijms21041452] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 02/13/2020] [Accepted: 02/17/2020] [Indexed: 02/08/2023] Open
Abstract
Hyperbaric oxygen (HBO) therapy has been reported to be beneficial for treating many conditions of inflammation-associated bone loss. The aim of this work was to in vitro investigate the effect of HBO in the course of osteogenesis of human Mesenchymal Stem Cells (MSCs) grown in a simulated pro-inflammatory environment. Cells were cultured with osteogenic differentiation factors in the presence or not of the pro-inflammatory cytokine Tumor Necrosis Factor-α (TNF-α), and simultaneously exposed daily for 60 min, and up to 21 days, at 2,4 atmosphere absolute (ATA) and 100% O2. To elucidate osteogenic differentiation-dependent effects, cells were additionally pre-committed prior to treatments. Cell metabolic activity was evaluated by means of the MTT assay and DNA content quantification, whereas osteogenic and vasculogenic differentiation was assessed by quantification of extracellular calcium deposition and gene expression analysis. Metabolic activity and osteogenic properties of cells did not differ between HBO, high pressure (HB) alone, or high oxygen (HO) alone and control if cells were pre-differentiated to the osteogenic lineage. In contrast, when treatments started contextually to the osteogenic differentiation of the cells, a significant reduction in cell metabolic activity first, and in mineral deposition at later time points, were observed in the HBO-treated group. Interestingly, TNF-α supplementation determined a significant improvement in the osteogenic capacity of cells subjected to HBO, which was not observed in TNF-α-treated cells exposed to HB or HO alone. This study suggests that exposure of osteogenic-differentiating MSCs to HBO under in vitro simulated inflammatory conditions enhances differentiation towards the osteogenic phenotype, providing evidence of the potential application of HBO in all those processes requiring bone regeneration.
Collapse
Affiliation(s)
- Chiara Gardin
- Maria Cecilia Hospital, GVM Care & Research, 48033 Cotignola (RA), Italy; (C.G.); (L.F.)
- Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy
| | - Gerardo Bosco
- Department of Biomedical Sciences, University of Padova, 35128 Padova, Italy; (G.B.); (S.Q.); (A.R.)
| | - Letizia Ferroni
- Maria Cecilia Hospital, GVM Care & Research, 48033 Cotignola (RA), Italy; (C.G.); (L.F.)
- Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy
| | - Silvia Quartesan
- Department of Biomedical Sciences, University of Padova, 35128 Padova, Italy; (G.B.); (S.Q.); (A.R.)
| | - Alex Rizzato
- Department of Biomedical Sciences, University of Padova, 35128 Padova, Italy; (G.B.); (S.Q.); (A.R.)
| | - Marco Tatullo
- Department of Basic Medical Sciences, Neurosciences and Sense Organs, University of Bari “Aldo Moro”, 70121 Bari, Italy
- Correspondence: (B.Z.); (M.T.); Tel.: +39-0532-455-502 (B.Z.)
| | - Barbara Zavan
- Maria Cecilia Hospital, GVM Care & Research, 48033 Cotignola (RA), Italy; (C.G.); (L.F.)
- Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy
- Correspondence: (B.Z.); (M.T.); Tel.: +39-0532-455-502 (B.Z.)
| |
Collapse
|
40
|
Chiesa I, De Maria C, Lapomarda A, Fortunato GM, Montemurro F, Di Gesù R, Tuan RS, Vozzi G, Gottardi R. Endothelial cells support osteogenesis in an in vitro vascularized bone model developed by 3D bioprinting. Biofabrication 2020; 12:025013. [PMID: 31929117 DOI: 10.1088/1758-5090/ab6a1d] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Bone is a highly vascularized tissue, in which vascularization and mineralization are concurrent processes during skeletal development. Indeed, both components should be included in any reliable and adherent in vitro model platform for the study of bone physiology and pathogenesis of skeletal disorders. To this end, we developed an in vitro vascularized bone model, using a gelatin-nanohydroxyapatite (gel-nHA) three-dimensional (3D) bioprinted scaffold. First, we seeded human mesenchymal stem cells (hMSCs) on the scaffold, which underwent osteogenic differentiation for 2 weeks. Then, we included lentiviral-GFP transfected human umbilical vein endothelial cells (HUVECs) within the 3D bioprinted scaffold macropores to form a capillary-like network during 2 more weeks of culture. We tested three experimental conditions: condition 1, bone constructs with HUVECs cultured in 1:1 osteogenic medium (OM): endothelial medium (EM); condition 2, bone constructs without HUVECs cultured in 1:1 OM:EM; condition 3: bone construct with HUVECs cultured in 1:1 growth medium:EM. All samples resulted in engineered bone matrix. In conditions 1 and 3, HUVECs formed tubular structures within the bone constructs, with the assembly of a complex capillary-like network visible by fluorescence microscopy in the live tissue and histology. CD31 immunostaining confirmed significant vascular lumen formation. Quantitative real-time PCR was used to quantify osteogenic differentiation and endothelial response. Alkaline phosphatase and runt-related transcription factor 2 upregulation confirmed early osteogenic commitment of hMSCs. Even when OM was removed under condition 3, we observed clear osteogenesis, which was notably accompanied by upregulation of osteopontin, vascular endothelial growth factor, and collagen type I. These findings indicate that we have successfully realized a bone model with robust vascularization in just 4 weeks of culture and we highlighted how the inclusion of endothelial cells more realistically supports osteogenesis. The approach reported here resulted in a biologically inspired in vitro model of bone vascularization, simulating de novo morphogenesis of capillary vessels occurring during tissue development.
Collapse
Affiliation(s)
- Irene Chiesa
- Research Center 'E. Piaggio', University of Pisa, Pisa, Italy. Center for Cellular and Molecular Engineering, Department of Orthopedic Surgery, School of Medicine, University of Pittsburgh, Pittsburgh, United States of America. Department of Pediatrics, Division of Pulmonary Medicine, The Children's Hospital of Philadelphia, Philadelphia, United States of America. Dept. of Ingegneria dell'Informazione, University of Pisa, Pisa, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Shahabipour F, Ashammakhi N, Oskuee RK, Bonakdar S, Hoffman T, Shokrgozar MA, Khademhosseini A. Key components of engineering vascularized 3-dimensional bioprinted bone constructs. Transl Res 2020; 216:57-76. [PMID: 31526771 DOI: 10.1016/j.trsl.2019.08.010] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2019] [Revised: 08/28/2019] [Accepted: 08/30/2019] [Indexed: 12/16/2022]
Abstract
Vascularization has a pivotal role in engineering successful tissue constructs. However, it remains a major hurdle of bone tissue engineering, especially in clinical applications for the treatment of large bone defects. Development of vascularized and clinically-relevant engineered bone substitutes with sufficient blood supply capable of maintaining implant viability and supporting subsequent host tissue integration remains a major challenge. Since only cells that are 100-200 µm from blood vessels can receive oxygen through diffusion, engineered constructs that are thicker than 400 µm face a challenging oxygenation problem. Following implantation in vivo, spontaneous ingrowth of capillaries in thick engineered constructs is too slow. Thus, it is critical to provide optimal conditions to support vascularization in engineered bone constructs. To achieve this, an in-depth understanding of the mechanisms of angiogenesis and bone development is required. In addition, it is also important to mimic the physiological milieu of native bone to fabricate more successful vascularized bone constructs. Numerous applications of engineered vascularization with cell-and/or microfabrication-based approaches seek to meet these aims. Three-dimensional (3D) printing promises to create patient-specific bone constructs in the future. In this review, we discuss the major components of fabricating vascularized 3D bioprinted bone constructs, analyze their related challenges, and highlight promising future trends.
Collapse
Affiliation(s)
- Fahimeh Shahabipour
- National cell bank of Iran, Pasteur Institute of Iran, Tehran, Iran; Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, Los Angeles, California; California NanoSystems Institute (CNSI), University of California, Los Angeles, Los Angeles, California; Department of Bioengineering, University of California, Los Angeles, Los Angeles, California
| | - Nureddin Ashammakhi
- Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, Los Angeles, California; California NanoSystems Institute (CNSI), University of California, Los Angeles, Los Angeles, California; Department of Bioengineering, University of California, Los Angeles, Los Angeles, California; Department of Radiological Sciences, University of California, Los Angeles, Los Angeles, California
| | - Reza K Oskuee
- Targeted Drug Delivery Research Center, Institute of Pharmaceutical Technology, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Shahin Bonakdar
- National cell bank of Iran, Pasteur Institute of Iran, Tehran, Iran
| | - Tyler Hoffman
- Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, Los Angeles, California; California NanoSystems Institute (CNSI), University of California, Los Angeles, Los Angeles, California; Department of Bioengineering, University of California, Los Angeles, Los Angeles, California
| | | | - Ali Khademhosseini
- Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, Los Angeles, California; California NanoSystems Institute (CNSI), University of California, Los Angeles, Los Angeles, California; Department of Bioengineering, University of California, Los Angeles, Los Angeles, California; Department of Radiological Sciences, University of California, Los Angeles, Los Angeles, California; Department of Chemical and Biomolecular Engineering, University of California, Los Angeles, Los Angeles, California.
| |
Collapse
|
42
|
Stefanowski J, Lang A, Rauch A, Aulich L, Köhler M, Fiedler AF, Buttgereit F, Schmidt-Bleek K, Duda GN, Gaber T, Niesner RA, Hauser AE. Spatial Distribution of Macrophages During Callus Formation and Maturation Reveals Close Crosstalk Between Macrophages and Newly Forming Vessels. Front Immunol 2019; 10:2588. [PMID: 31956322 PMCID: PMC6953593 DOI: 10.3389/fimmu.2019.02588] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 10/18/2019] [Indexed: 01/15/2023] Open
Abstract
Macrophages are essential players in the process of fracture healing, acting by remodeling of the extracellular matrix and enabling vascularization. Whilst activated macrophages of M1-like phenotype are present in the initial pro-inflammatory phase of hours to days of fracture healing, an anti-inflammatory M2-like macrophage phenotype is supposed to be crucial for the induction of downstream cascades of healing, especially the initiation of vascularization. In a mouse-osteotomy model, we provide a comprehensive characterization of vessel (CD31+, Emcn+) and macrophage phenotypes (F4/80, CD206, CD80, Mac-2) during the process of fracture healing. To this end, we phenotype the phases of vascular regeneration-the expansion phase (d1-d7 after injury) and the remodeling phase of the endothelial network, until tissue integrity is restored (d14-d21 after injury). Vessels which appear during the bone formation process resemble type H endothelium (CD31hiEmcnhi), and are closely connected to osteoprogenitors (Runx2+, Osx+) and F4/80+ macrophages. M1-like macrophages are present in the initial phase of vascularization until day 3 post osteotomy, but they are rare during later regeneration phases. M2-like macrophages localize mainly extramedullary, and CD206+ macrophages are found to express Mac-2+ during the expansion phase. VEGFA expression is initiated by CD80+ cells, including F4/80+ macrophages, until day 3, while subsequently osteoblasts and chondrocytes are main contributors to VEGFA production at the fracture site. Using Longitudinal Intravital Microendoscopy of the Bone (LIMB) we observe changes in the motility and organization of CX3CR1+ cells, which infiltrate the injury site after an osteotomy. A transient accumulation, resulting in spatial polarization of both, endothelial cells and macrophages, in regions distal to the fracture site, is evident. Immunofluorescence histology followed by histocytometric analysis reveals that F4/80+CX3CR1+ myeloid cells precede vascularization.
Collapse
Affiliation(s)
- Jonathan Stefanowski
- Department of Rheumatology and Clinical Immunology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,German Rheumatism Research Centre (DRFZ) Berlin, a Leibniz Institute, Berlin, Germany
| | - Annemarie Lang
- Department of Rheumatology and Clinical Immunology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,German Rheumatism Research Centre (DRFZ) Berlin, a Leibniz Institute, Berlin, Germany.,Berlin-Brandenburg Center for Regenerative Therapies, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Ariana Rauch
- German Rheumatism Research Centre (DRFZ) Berlin, a Leibniz Institute, Berlin, Germany
| | - Linus Aulich
- German Rheumatism Research Centre (DRFZ) Berlin, a Leibniz Institute, Berlin, Germany
| | - Markus Köhler
- German Rheumatism Research Centre (DRFZ) Berlin, a Leibniz Institute, Berlin, Germany
| | - Alexander F Fiedler
- German Rheumatism Research Centre (DRFZ) Berlin, a Leibniz Institute, Berlin, Germany
| | - Frank Buttgereit
- Department of Rheumatology and Clinical Immunology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,German Rheumatism Research Centre (DRFZ) Berlin, a Leibniz Institute, Berlin, Germany.,Berlin-Brandenburg Center for Regenerative Therapies, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Katharina Schmidt-Bleek
- Berlin-Brandenburg Center for Regenerative Therapies, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,Julius Wolff Institute for Biomechanics and Musculoskeletal Regeneration, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Georg N Duda
- Berlin-Brandenburg Center for Regenerative Therapies, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,Julius Wolff Institute for Biomechanics and Musculoskeletal Regeneration, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Timo Gaber
- Department of Rheumatology and Clinical Immunology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,German Rheumatism Research Centre (DRFZ) Berlin, a Leibniz Institute, Berlin, Germany.,Berlin-Brandenburg Center for Regenerative Therapies, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Raluca A Niesner
- German Rheumatism Research Centre (DRFZ) Berlin, a Leibniz Institute, Berlin, Germany.,Dynamic and Functional in vivo Imaging, Department of Veterinary Medicine, Freie Universität Berlin, Berlin, Germany
| | - Anja E Hauser
- Department of Rheumatology and Clinical Immunology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,German Rheumatism Research Centre (DRFZ) Berlin, a Leibniz Institute, Berlin, Germany
| |
Collapse
|
43
|
Hadida M, Marchat D. Strategy for achieving standardized bone models. Biotechnol Bioeng 2019; 117:251-271. [PMID: 31531968 PMCID: PMC6915912 DOI: 10.1002/bit.27171] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 09/05/2019] [Accepted: 09/06/2019] [Indexed: 12/24/2022]
Abstract
Reliably producing functional in vitro organ models, such as organ-on-chip systems, has the potential to considerably advance biology research, drug development time, and resource efficiency. However, despite the ongoing major progress in the field, three-dimensional bone tissue models remain elusive. In this review, we specifically investigate the control of perfusion flow effects as the missing link between isolated culture systems and scientifically exploitable bone models and propose a roadmap toward this goal.
Collapse
Affiliation(s)
- Mikhael Hadida
- Mines Saint-Etienne, Univ Lyon, Univ Jean Monnet, INSERM, U 1059 Sainbiose, Centre CIS, Saint-Etienne, France
| | - David Marchat
- Mines Saint-Etienne, Univ Lyon, Univ Jean Monnet, INSERM, U 1059 Sainbiose, Centre CIS, Saint-Etienne, France
| |
Collapse
|
44
|
Human Umbilical Vein Endothelial Cells (HUVECs) Co-Culture with Osteogenic Cells: From Molecular Communication to Engineering Prevascularised Bone Grafts. J Clin Med 2019; 8:jcm8101602. [PMID: 31623330 PMCID: PMC6832897 DOI: 10.3390/jcm8101602] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 09/12/2019] [Accepted: 09/23/2019] [Indexed: 12/21/2022] Open
Abstract
The repair of bone defects caused by trauma, infection or tumor resection is a major clinical orthopedic challenge. The application of bone grafts in orthopedic procedures is associated with a problem of inadequate vascularization in the initial phase after implantation. Meanwhile, the survival of cells within the implanted graft and its integration with the host tissue is strongly dependent on nutrient and gaseous exchange, as well as waste product removal, which are effectuated by blood microcirculation. In the bone tissue, the vasculature also delivers the calcium and phosphate indispensable for the mineralization process. The critical role of vascularization for bone healing and function, led the researchers to the idea of generating a capillary-like network within the bone graft in vitro, which could allow increasing the cell survival and graft integration with a host tissue. New strategies for engineering pre-vascularized bone grafts, that apply the co-culture of endothelial and bone-forming cells, have recently gained interest. However, engineering of metabolically active graft, containing two types of cells requires deep understanding of the underlying mechanisms of interaction between these cells. The present review focuses on the best-characterized endothelial cells-human umbilical vein endothelial cells (HUVECs)-attempting to estimate whether the co-culture approach, using these cells, could bring us closer to development and possible clinical application of prevascularized bone grafts.
Collapse
|
45
|
Hou W, Xia J, Liu C, Li S, Wu T, Huang Y. Development of a method to screen and isolate bioactive constituents from Stellera chamaejasme by ultrafiltration and liquid chromatography combined with semi-preparative high-performance liquid chromatography and high-speed counter current chromatography. J Sep Sci 2019; 42:3421-3431. [PMID: 31529668 DOI: 10.1002/jssc.201900772] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 08/27/2019] [Accepted: 09/11/2019] [Indexed: 12/20/2022]
Abstract
A simple and efficient method based on ultrafiltration with liquid chromatography and mass spectrometry was used for the rapid screening and identification of ligands in the extracts of Stellera chamaejasme. The bound ligands, i.e. daphnoretin, isopimpinellin, chamaechromone, neochamaejasmin A, and chamaejasmine (purity of 96.8, 90.75, 91.41, 93.98, and 98.91%, respectively), were separated by semi-preparative high-performance liquid chromatography combined with high-speed counter-current chromatography. To the best of our knowledge, this is the first study to report the detection of potent lipoxidase and lactate dehydrogenase inhibitors in Stellera chamaejasme extracts. The results demonstrate that our method of ultrafiltration with liquid chromatography and mass spectrometry combined with mixed chromatography can be used to screen and confirm the bioactivity of all isolated compounds. This method also eliminates the need for separation of inactive compounds, thereby improving efficiency when studying bioactive substances. For some complex mixtures, neither semi-preparative high-performance liquid chromatography nor high-speed counter-current chromatography can purify all the target active compounds with high purity in a one-step separation. The combination of the two methods allow for efficient purification of target bioactive compounds with different polarities and physicochemical properties based on their complementary properties.
Collapse
Affiliation(s)
- Wanchao Hou
- Central Laboratory, Changchun Normal University, Changchun, P. R. China
| | - Jianli Xia
- Central Laboratory, Changchun Normal University, Changchun, P. R. China
| | - Chunming Liu
- Central Laboratory, Changchun Normal University, Changchun, P. R. China
| | - Sainan Li
- Central Laboratory, Changchun Normal University, Changchun, P. R. China
| | - Tong Wu
- Central Laboratory, Changchun Normal University, Changchun, P. R. China
| | - Yu Huang
- Central Laboratory, Changchun Normal University, Changchun, P. R. China
| |
Collapse
|
46
|
Synthesis and characterization of CaSr-Metal Organic Frameworks for biodegradable orthopedic applications. Sci Rep 2019; 9:13024. [PMID: 31506530 PMCID: PMC6736967 DOI: 10.1038/s41598-019-49536-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Accepted: 08/23/2019] [Indexed: 12/20/2022] Open
Abstract
Metal-organic frameworks (MOFs) formed from metals and organic ligands, are crystalline materials that are degradable in aqueous medium, and capable of releasing Ca and Sr ions. In this manuscript, the ability of MOFs to degrade and release osteogenic Ca and Sr ions was investigated. MOFs were generated by choosing osteoinductive Ca and Sr metals, and an organic ligand 1,3,5 tricarboxylicbenzene (H3BTC) as a linker. These MOFs were able to induce in vitro biomineralization from pre-osteoblastic MC3T3 cells and human mesenchymal stem cells (hMSCs). Moreover, these MOFs (when loaded with dimethyloxalylglycine (DMOG)) induced vascular endothelial production from hMSCs. qRT-PCR analysis performed on hMSCs (isolated from femoral heads of patients undergoing joint arthroplasty) treated with MOFs crystals suggested that the CaSr-MOFs by themselves can upregulate osteogenic genes in hMSCs, which is the first time to our knowledge that this has been observed from MOFs.
Collapse
|
47
|
Clumps of Mesenchymal Stem Cell/Extracellular Matrix Complexes Generated with Xeno-Free Conditions Facilitate Bone Regeneration via Direct and Indirect Osteogenesis. Int J Mol Sci 2019; 20:ijms20163970. [PMID: 31443173 PMCID: PMC6720767 DOI: 10.3390/ijms20163970] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 08/09/2019] [Accepted: 08/12/2019] [Indexed: 02/06/2023] Open
Abstract
Three-dimensional clumps of mesenchymal stem cell (MSC)/extracellular matrix (ECM) complexes (C-MSCs) consist of cells and self-produced ECM. We demonstrated previously that C-MSCs can be transplanted into bone defect regions with no artificial scaffold to induce bone regeneration. To apply C-MSCs in a clinical setting as a reliable bone regenerative therapy, the present study aimed to generate C-MSCs in xeno-free/serum-free conditions that can exert successful bone regenerative properties and to monitor interactions between grafted cells and host cells during bone healing processes. Human bone marrow-derived MSCs were cultured in xeno-free/serum-free medium. To obtain C-MSCs, confluent cells that had formed on the cellular sheet were scratched using a micropipette tip and then torn off. The sheet was rolled to make a round clump of cells. Then, C-MSCs were transplanted into an immunodeficient mouse calvarial defect model. Transplantation of C-MSCs induced bone regeneration in a time-dependent manner. Immunofluorescence staining showed that both donor human cells and host mice cells contributed to bone reconstruction. Decellularized C-MSCs implantation failed to induce bone regeneration, even though the host mice cells can infiltrate into the defect area. These findings suggested that C-MSCs generated in xeno-free/serum-free conditions can induce bone regeneration via direct and indirect osteogenesis.
Collapse
|
48
|
Abstract
Human bones have unique structures and characteristics, and replacing a natural bone in the case of bone fracture or bone diseases is a very complicated problem. The main goal of this paper was to summarize the recent research on polymer materials as bone substitutes and for bone repair. Bone treatment methods, bone substitute materials as well as their advantages and drawbacks, and manufacturing methods were reviewed. Biopolymers are the most promising materials in the field of artificial bones and using biopolymers with the shape memory effect can improve the integration of an artificial bone into the human body by better mimicking the structure and properties of natural bones, decreasing the invasiveness of surgical procedures by producing deployable implants. It has been shown that the application of the rapid prototyping technology for artificial bones allows the customization of bone substitutes for a patient and the creation of artificial bones with a complex structure.
Collapse
Affiliation(s)
- Anastasiia Kashirina
- Department of Astronautical Science and Mechanics, Harbin Institute of Technology, PO Box 301, No. 92 West Dazhi Street, Harbin 150001, China
| | - Yongtao Yao
- National Key Laboratory of Science and Technology on Advanced Composites in Special Environments, Harbin Institute of Technology, No. 2 YiKuang Street, Harbin 150080, China.
| | - Yanju Liu
- Department of Astronautical Science and Mechanics, Harbin Institute of Technology, PO Box 301, No. 92 West Dazhi Street, Harbin 150001, China
| | - Jinsong Leng
- National Key Laboratory of Science and Technology on Advanced Composites in Special Environments, Harbin Institute of Technology, No. 2 YiKuang Street, Harbin 150080, China.
| |
Collapse
|
49
|
Piluso S, Li Y, Abinzano F, Levato R, Moreira Teixeira L, Karperien M, Leijten J, van Weeren R, Malda J. Mimicking the Articular Joint with In Vitro Models. Trends Biotechnol 2019; 37:1063-1077. [PMID: 31000204 DOI: 10.1016/j.tibtech.2019.03.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 02/21/2019] [Accepted: 03/04/2019] [Indexed: 12/18/2022]
Abstract
Treating joint diseases remains a significant clinical challenge. Conventional in vitro cultures and animal models have been helpful, but suffer from limited predictive power for the human response. Advanced models are therefore required to mimic the complex biological interactions within the human joint. However, the intricate structure of the joint microenvironment and the complex nature of joint diseases have challenged the development of in vitro models that can faithfully mimic the in vivo physiological and pathological environments. In this review, we discuss the current in vitro models of the joint and the progress achieved in the development of novel and potentially more predictive models, and highlight the application of new technologies to accurately emulate the articular joint.
Collapse
Affiliation(s)
- Susanna Piluso
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands; Department of Developmental BioEngineering, Technical Medical Centre, University of Twente, Enschede, The Netherlands; Regenerative Medicine Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Yang Li
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands; Regenerative Medicine Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Florencia Abinzano
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands; Regenerative Medicine Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Riccardo Levato
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands; Regenerative Medicine Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Liliana Moreira Teixeira
- Department of Developmental BioEngineering, Technical Medical Centre, University of Twente, Enschede, The Netherlands; Regenerative Medicine Utrecht, Utrecht University, Utrecht, The Netherlands; Department of Equine Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Marcel Karperien
- Department of Developmental BioEngineering, Technical Medical Centre, University of Twente, Enschede, The Netherlands
| | - Jeroen Leijten
- Department of Developmental BioEngineering, Technical Medical Centre, University of Twente, Enschede, The Netherlands
| | - René van Weeren
- Regenerative Medicine Utrecht, Utrecht University, Utrecht, The Netherlands; Department of Equine Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Jos Malda
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands; Regenerative Medicine Utrecht, Utrecht University, Utrecht, The Netherlands; Department of Equine Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands.
| |
Collapse
|
50
|
Fani N, Farokhi M, Azami M, Kamali A, Bakhshaiesh NL, Ebrahimi-Barough S, Ai J, Eslaminejad MB. Endothelial and Osteoblast Differentiation of Adipose-Derived Mesenchymal Stem Cells Using a Cobalt-Doped CaP/Silk Fibroin Scaffold. ACS Biomater Sci Eng 2019; 5:2134-2146. [DOI: 10.1021/acsbiomaterials.8b01372] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Nesa Fani
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, 1417755469 Tehran, Iran
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, 1665659911, ACECR, Tehran, Iran
| | - Mehdi Farokhi
- National Cell Bank of Iran, Pasteur Institute of Iran, 1316943551 Tehran, Iran
| | - Mahmoud Azami
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, 1417755469 Tehran, Iran
| | - Amir Kamali
- Department of Pathology, School of Veterinary Medicine, Shiraz University, 7194684471 Shiraz, Iran
| | - Nasrin Lotfi Bakhshaiesh
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, 1417755469 Tehran, Iran
| | - Somayeh Ebrahimi-Barough
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, 1417755469 Tehran, Iran
| | - Jafar Ai
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, 1417755469 Tehran, Iran
| | - Mohamadreza Baghaban Eslaminejad
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, 1665659911, ACECR, Tehran, Iran
| |
Collapse
|