1
|
Arash A, Dehgan F, Zamanlui Benisi S, Jafari-Nodoushan M, Pezeshki-Modaress M. Polysaccharide base electrospun nanofibrous scaffolds for cartilage tissue engineering: Challenges and opportunities. Int J Biol Macromol 2024; 277:134054. [PMID: 39038580 DOI: 10.1016/j.ijbiomac.2024.134054] [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: 05/15/2024] [Revised: 07/12/2024] [Accepted: 07/18/2024] [Indexed: 07/24/2024]
Abstract
Polysaccharides, known as naturally abundant macromolecular materials which can be easily modified chemically, have always attracted scientists' interest due to their outstanding properties in tissue engineering. Moreover, their intrinsic similarity to cartilage ECM components, biocompatibility, and non-harsh processing conditions make polysaccharides an excellent option for cartilage tissue engineering. Imitating the natural ECM structure to form a fibrous scaffold at the nanometer scale in order to recreate the optimal environment for cartilage regeneration has always been attractive for researchers in the past few years. However, there are some challenges for polysaccharides electrospun nanofibers preparation, such as poor solubility (Alginate, cellulose, chitin), high viscosity (alginate, chitosan, and Hyaluronic acid), high surface tension, etc. Several methods are reported in the literature for facing polysaccharide electrospinning issues, such as using carrier polymers, modification of polysaccharides, and using different solvent systems. In this review, considering the importance of polysaccharide-based electrospun nanofibers in cartilage tissue engineering applications, the main achievements in the past few years, and challenges for their electrospinning process are discussed. After careful investigation of reported studies in the last few years, alginate, chitosan, hyaluronic acid, chondroitin sulfate, and cellulose were chosen as the main polysaccharide base electrospun nanofibers used for cartilage regeneration.
Collapse
Affiliation(s)
- Atefeh Arash
- Department of Biomedical Engineering, Faculty of Engineering, Islamic Azad University, Central Tehran Branch, Tehran, Iran
| | - Fatemeh Dehgan
- Department of Biomedical Engineering, Faculty of Engineering, Islamic Azad University, Central Tehran Branch, Tehran, Iran
| | - Soheila Zamanlui Benisi
- Department of Biomedical Engineering, Islamic Azad University, Central Tehran Branch, Tehran, Iran; Stem cells Research Center, Tissue Engineering and Regenerative Medicine Institute, Islamic Azad University, Central Tehran Branch, Tehran, Iran
| | - Milad Jafari-Nodoushan
- Department of Biomedical Engineering, Islamic Azad University, Central Tehran Branch, Tehran, Iran; Hard Tissue Engineering Resarch Center, Tissue Engineering and Regenerative Medicine Institute, Central Tehran Branch, Islamic Azad University, Tehran, Iran.
| | - Mohamad Pezeshki-Modaress
- Burn Research Center, Iran University of Medical Sciences, Tehran, Iran; Department of Plastic and Reconstructive surgery, Hazrat Fatemeh Hospital, School of Medicine, Iran University of Medical Sciences, Tehran, Iran; Stem Cell and Regenerative Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
2
|
Olate-Moya F, Rubí-Sans G, Engel E, Mateos-Timoneda MÁ, Palza H. 3D Bioprinting of Biomimetic Alginate/Gelatin/Chondroitin Sulfate Hydrogel Nanocomposites for Intrinsically Chondrogenic Differentiation of Human Mesenchymal Stem Cells. Biomacromolecules 2024; 25:3312-3324. [PMID: 38728671 DOI: 10.1021/acs.biomac.3c01444] [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: 05/12/2024]
Abstract
3D-printed hydrogel scaffolds biomimicking the extracellular matrix (ECM) are key in cartilage tissue engineering as they can enhance the chondrogenic differentiation of mesenchymal stem cells (MSCs) through the presence of active nanoparticles such as graphene oxide (GO). Here, biomimetic hydrogels were developed by cross-linking alginate, gelatin, and chondroitin sulfate biopolymers in the presence of GO as a bioactive filler, with excellent processability for developing bioactive 3D printed scaffolds and for the bioprinting process. A novel bioink based on our hydrogel with embedded human MSCs presented a cell survival rate near 100% after the 3D bioprinting process. The effects of processing and filler concentration on cell differentiation were further quantitatively evaluated. The nanocomposited hydrogels render high MSC proliferation and viability, exhibiting intrinsic chondroinductive capacity without any exogenous factor when used to print scaffolds or bioprint constructs. The bioactivity depended on the GO concentration, with the best performance at 0.1 mg mL-1. These results were explained by the rational combination of the three biopolymers, with GO nanoparticles having carboxylate and sulfate groups in their structures, therefore, biomimicking the highly negatively charged ECM of cartilage. The bioactivity of this biomaterial and its good processability for 3D printing scaffolds and 3D bioprinting techniques open up a new approach to developing novel biomimetic materials for cartilage repair.
Collapse
Affiliation(s)
- Felipe Olate-Moya
- Departamento de Ingeniería Química, Biotecnología y Materiales, Facultad de Ciencias Físicas y Matemáticas, Universidad de Chile, Avenida Beauchef 851, 8370458 Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Avenida Monseñor Álvaro del Portillo 12455, 7620086 Las Condes, Chile
| | - Gerard Rubí-Sans
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Carrer de Baldiri Reixac, 10, 12, 08028, 08019 Barcelona, Spain
- CIBER en Bioingeniería, Biomateriales y Nanomedicina, CIBER-BBN, 50018 Zaragoza, Spain
| | - Elisabeth Engel
- IMEM-BRT Group, Departament de Ciència i Enginyeria de Materials, EEBE, Universitat Politècnica de Catalunya (UPC), C/Eduard Maristany 10-14, 08019 Barcelona, Spain
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Carrer de Baldiri Reixac, 10, 12, 08028, 08019 Barcelona, Spain
- CIBER en Bioingeniería, Biomateriales y Nanomedicina, CIBER-BBN, 50018 Zaragoza, Spain
| | - Miguel Ángel Mateos-Timoneda
- Bioengineering Institute of Technology, Universitat Internacional de Catalunya, Josep Trueta Street s/n, 08195 Sant Cugat del Vallès, Barcelona, Spain
- Department of Basic Sciences, Faculty of Medicine and Health Sciences, Univesitat Internacional de Catalunya, Josep Trueta Street s/n, 08195 Sant Cugat del Vallès, Barcelona, Spain
| | - Humberto Palza
- Departamento de Ingeniería Química, Biotecnología y Materiales, Facultad de Ciencias Físicas y Matemáticas, Universidad de Chile, Avenida Beauchef 851, 8370458 Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Avenida Monseñor Álvaro del Portillo 12455, 7620086 Las Condes, Chile
| |
Collapse
|
3
|
Qi Y, Lv H, Huang Q, Pan G. The Synergetic Effect of 3D Printing and Electrospinning Techniques in the Fabrication of Bone Scaffolds. Ann Biomed Eng 2024; 52:1518-1533. [PMID: 38530536 DOI: 10.1007/s10439-024-03500-5] [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: 02/07/2024] [Accepted: 03/19/2024] [Indexed: 03/28/2024]
Abstract
The primary goal of bone tissue engineering is to restore and rejuvenate bone defects by using a suitable three-dimensional scaffold, appropriate cells, and growth hormones. Various scaffolding methods are used to fabricate three-dimensional scaffolds, which provide the necessary environment for cell activity and bone formation. Multiple materials may be used to create scaffolds with hierarchical structures that are optimal for cell growth and specialization. This study examines a notion for creating an optimal framework for bone regeneration using a combination of the robocasting method and the electrospinning approach. Research indicates that the integration of these two procedures enhances the benefits of each method and provides a rationale for addressing their shortcomings via this combination. The hybrid approach is anticipated to provide a manufactured scaffold that can effectively replace bone defects while possessing the necessary qualities for bone regeneration.
Collapse
Affiliation(s)
- Yongjie Qi
- School of Intelligent Manufacturing, Zhejiang Guangsha Vocational and Technical University of Construction, Dongyang, 322100, China
| | - Hangying Lv
- School of Intelligent Manufacturing, Zhejiang Guangsha Vocational and Technical University of Construction, Dongyang, 322100, China
| | - Qinghua Huang
- School of Intelligent Manufacturing, Zhejiang Guangsha Vocational and Technical University of Construction, Dongyang, 322100, China
| | - Guangyong Pan
- School of Intelligent Manufacturing, Zhejiang Guangsha Vocational and Technical University of Construction, Dongyang, 322100, China.
| |
Collapse
|
4
|
Noh S, Jin YJ, Shin DI, Kwon HJ, Yun HW, Kim KM, Park JY, Chung JY, Park DY. Selective Extracellular Matrix Guided Mesenchymal Stem Cell Self-Aggregate Engineering for Replication of Meniscal Zonal Tissue Gradient in a Porcine Meniscectomy Model. Adv Healthc Mater 2023; 12:e2301180. [PMID: 37463568 DOI: 10.1002/adhm.202301180] [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: 04/14/2023] [Revised: 06/13/2023] [Accepted: 07/14/2023] [Indexed: 07/20/2023]
Abstract
Degenerative meniscus tears (DMTs) are prevalent findings in osteoarthritic knees, yet current treatment is mostly limited to arthroscopic partial meniscectomy rather than regeneration, which further exacerbates arthritic changes. Translational research regarding meniscus regeneration is hindered by the complex, composite nature of the meniscus which exhibit a gradient from inner cartilage-like tissue to outer fibrous tissue, as well as engineering hurdles often requiring growth factors and cross-linking agents. Here, a meniscus zonal tissue gradient is proposed using zone-specific decellularized meniscus extracellular matrix (DMECM) and autologous synovial mesenchymal stem cells (SMSC) via self-aggregation without the use of growth factors or cross-linking agents. Combination with zone-specific DMECM during self-aggregation of MSCs forms zone-specific meniscus tissue that reflects the respective DMECM harvest site. The implantation of these constructs leads to the regeneration of meniscus tissue resembling the native meniscus, demonstrating inner cartilaginous and outer fibrous characteristics as well as recovery of native meniscal microarchitecture in a porcine partial meniscectomy model at 6 months. In all, the findings offer a potential regenerative therapy for DMTs that may improve current partial meniscectomy-based patient care.
Collapse
Affiliation(s)
- Sujin Noh
- Department of Biomedical Sciences, Graduate School of Ajou University, Suwon, 16499, Republic of Korea
| | - Yong Jun Jin
- Department of Orthopedic Surgery, School of Medicine, Ajou University, Suwon, 16499, Republic of Korea
| | - Dong Il Shin
- Department of Molecular Science and Technology, Ajou University, Suwon, 16499, Republic of Korea
| | - Hyeon Jae Kwon
- Department of Molecular Science and Technology, Ajou University, Suwon, 16499, Republic of Korea
| | - Hee-Woong Yun
- Department of Orthopedic Surgery, School of Medicine, Ajou University, Suwon, 16499, Republic of Korea
- Cell Therapy Center, Ajou Medical Center, Suwon, 16499, Republic of Korea
| | - Kyu Min Kim
- Cell Therapy Center, Ajou Medical Center, Suwon, 16499, Republic of Korea
| | - Jae-Young Park
- Department of Orthopedics Surgery, CHA University Bundang Medical Center, Bundang-gu, Seongnam-si, Gyeonggi-do, 13496, Republic of Korea
| | - Jun Young Chung
- Department of Orthopedic Surgery, School of Medicine, Ajou University, Suwon, 16499, Republic of Korea
| | - Do Young Park
- Department of Biomedical Sciences, Graduate School of Ajou University, Suwon, 16499, Republic of Korea
- Department of Orthopedic Surgery, School of Medicine, Ajou University, Suwon, 16499, Republic of Korea
- Cell Therapy Center, Ajou Medical Center, Suwon, 16499, Republic of Korea
- Ajou University, Leading Convergence of Healthcare and Medicine, Institute of Science & Technology (ALCHeMIST), Suwon, 16499, Republic of Korea
| |
Collapse
|
5
|
Kabirkoohian A, Bakhshi H, Irani S, Sharifi F. Chemical Immobilization of Carboxymethyl Chitosan on Polycaprolactone Nanofibers as Osteochondral Scaffolds. Appl Biochem Biotechnol 2023; 195:3888-3899. [PMID: 35488953 PMCID: PMC10203026 DOI: 10.1007/s12010-022-03916-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/14/2022] [Indexed: 12/30/2022]
Abstract
Carboxymethyl chitosan (CMC) as a bio-based osteochondral inductive material was chemically immobilized on the surface of polycaprolactone (PCL) nanofibers to fabricate scaffolds for osteochondral tissue engineering applications. The chemical immobilization process included the aminolysis of ester bonds and bonding of the primary amines with glutaraldehyde as a coupling agent. The SEM and FTIR results confirmed the successfulness of the CMC immobilization. The fabricated scaffolds presented cell viabilities of > 82% and supported the attachment and proliferation of the human bone marrow mesenchymal stem cells (hBM-MSCs). The CMC-immobilized scaffolds concentration dependently induced the diverse osteochondral differentiation pathways for the hBM-MSCs without using any external differential agents. According to the Alcian Blue and Alizarin Red staining and immunocytochemistry results, scaffolds with a higher content of CMC presented more chondro-inductivity and less osteoinductivity. Thus, the CMC-immobilized scaffolds can be employed as great potential candidates for osteochondral tissue engineering applications.
Collapse
Affiliation(s)
- Anita Kabirkoohian
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Hadi Bakhshi
- Department of Life Science and Bioprocesses, Fraunhofer Institute for Applied Polymer Research IAP, Geiselbergstraße 68, 14476, Potsdam-Golm, Germany.
| | - Shiva Irani
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Fereshteh Sharifi
- Hard Tissue Engineering Research Center, Tissue Engineering and Regenerative Medicine Institute, Central Tehran Branch, Islamic Azad University, Tehran, Iran
| |
Collapse
|
6
|
Jalandhra GK, Molley TG, Hung TT, Roohani I, Kilian KA. In situ formation of osteochondral interfaces through "bone-ink" printing in tailored microgel suspensions. Acta Biomater 2023; 156:75-87. [PMID: 36055612 DOI: 10.1016/j.actbio.2022.08.052] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 07/21/2022] [Accepted: 08/23/2022] [Indexed: 01/18/2023]
Abstract
Osteochondral tissue has a complex hierarchical structure spanning subchondral bone to articular cartilage. Biomaterials approaches to mimic and repair these interfaces have had limited success, largely due to challenges in fabricating composite hard-soft interfaces with living cells. Biofabrication approaches have emerged as attractive methods to form osteochondral analogues through additive assembly of hard and soft components. We have developed a unique printing platform that is able to integrate soft and hard materials concurrently through freeform printing of mineralized constructs within tunable microgel suspensions containing living cells. A library of microgels based on gelatin were prepared, where the stiffness of the microgels and a liquid "filler" phase can be tuned for bioprinting while simultaneously directing differentiation. Tuning microgel stiffness and filler content differentially directs chondrogenesis and osteogenesis within the same construct, demonstrating how this technique can be used to fabricate osteochondral interfaces in a single step. Printing of a rapidly setting calcium phosphate cement, so called "bone-ink" within a cell laden suspension bath further guides differentiation, where the cells adjacent to the nucleated hydroxyapatite phase undergo osteogenesis with cells in the surrounding medium undergoing chondrogenesis. In this way, bone analogues with hierarchical structure can be formed within cell-laden gradient soft matrices to yield multiphasic osteochondral constructs. This technique provides a versatile one-pot biofabrication approach without harsh post-processing which will aid efforts in bone disease modelling and tissue engineering. STATEMENT OF SIGNIFICANCE: This paper demonstrates the first example of a biofabrication approach to rapidly form osteochondral constructs in a single step under physiological conditions. Key to this advance is a tunable suspension of extracellular matrix microgels that are packed together with stem cells, providing a unique and modular scaffolding for guiding the simultaneous formation of bone and cartilage tissue. The physical properties of the suspension allow direct writing of a ceramic "bone-ink", resulting in an ordered structure of microscale hydrogels, living cells, and bone mimics in a single step. This platform reveals a simple approach to making complex skeletal tissue for disease modelling, with the possibility of repairing and replacing bone-cartilage interfaces in the clinic using a patient's own cells.
Collapse
Affiliation(s)
- Gagan K Jalandhra
- School of Materials Science and Engineering, University of New South Wales, Sydney NSW 2052; Australian Centre for NanoMedicine, University of New South Wales, Sydney NSW 2052
| | - Thomas G Molley
- School of Materials Science and Engineering, University of New South Wales, Sydney NSW 2052; Australian Centre for NanoMedicine, University of New South Wales, Sydney NSW 2052
| | - Tzong-Tyng Hung
- Biological Resources Imaging Laboratory, Mark Wainwright Analytical Centre, University of New South Wales, Sydney NSW 2052
| | - Iman Roohani
- School of Chemistry, University of New South Wales, Sydney NSW 2052; Australian Centre for NanoMedicine, University of New South Wales, Sydney NSW 2052
| | - Kristopher A Kilian
- School of Materials Science and Engineering, University of New South Wales, Sydney NSW 2052; School of Chemistry, University of New South Wales, Sydney NSW 2052; Australian Centre for NanoMedicine, University of New South Wales, Sydney NSW 2052.
| |
Collapse
|
7
|
Honarpardaz A, Daliri Joupari M, Tavakkoli S. In Vitro Chondrogenic Differentiation of Human Adipose-Derived Stem Cells by Diacerein. IRANIAN JOURNAL OF PHARMACEUTICAL RESEARCH : IJPR 2023; 22:e137803. [PMID: 38444710 PMCID: PMC10912900 DOI: 10.5812/ijpr-137803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 08/30/2023] [Accepted: 09/02/2023] [Indexed: 03/07/2024]
Abstract
Background Tissue engineering is the application system that tries to restore damaged tissues by different approaches, such as cellular therapy, application of cell differential factors, and various materials. One of the important goals in tissue engineering is to guide stem cells directly to the desired tissue, and researchers tried to utilize different molecules as effective factors to improve this technique. Objectives This study aims to demonstrate the effects of diacerein, a slow-acting drug for the treatment of osteoarthritis, on mesenchymal stem cell proliferation and evaluate its potential in the chondrogenesis process. Methods Stem cells were isolated from adipose tissue, characterized by flow cytometry, and cells were treated with 10-5M diacerein for three weeks. Chondrogenic gene expression of SOX9, COL2A1, ACAN, and TGFB1 were analyzed by qRT-PCR and immunocytochemistry techniques. Results Our results showed that diacerein increased the expression of the following genes involved in chondrogenesis: SOX9 (2.9-fold, P < 0.00), COL2A1 (2.2-fold, P < 0.00), ACAN (2.7-fold, P < 0.00), and TGFB1 (2.6-fold, P < 0.00). Immunocytochemistry results also showed increased production of collagen type II as the main protein marker for chondrocytes. Conclusions We observed that diacerein alone could initiate and enhance chondrogenesis, and it can be used as a differentiation factor for stem cells to chondrocyte besides its ability to inhibit IL-1β. Knowing the actual function of diacerein, it could be a good candidate for the treatment of osteoarthritis.
Collapse
Affiliation(s)
- Ali Honarpardaz
- Department of Animal Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | - Morteza Daliri Joupari
- Department of Animal Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | - Sajjad Tavakkoli
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
8
|
Um SH, Seo Y, Seo H, Lee K, Park SH, Jeon JH, Lim JY, Ok MR, Kim YC, Kim H, Cheon CH, Han HS, Edwards JR, Kim SW, Jeon H. Biomimetic hydrogel blanket for conserving and recovering intrinsic cell properties. Biomater Res 2022; 26:78. [PMID: 36514131 PMCID: PMC9746181 DOI: 10.1186/s40824-022-00327-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Accepted: 11/20/2022] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Cells in the human body experience different growth environments and conditions, such as compressive pressure and oxygen concentrations, depending on the type and location of the tissue. Thus, a culture device that emulates the environment inside the body is required to study cells outside the body. METHODS A blanket-type cell culture device (Direct Contact Pressing: DCP) was fabricated with an alginate-based hydrogel. Changes in cell morphology due to DCP pressure were observed using a phase contrast microscope. The changes in the oxygen permeability and pressure according to the hydrogel concentration of DCP were analyzed. To compare the effects of DCP with normal or artificial hypoxic cultures, cells were divided based on the culture technique: normal culture, DCP culture device, and artificial hypoxic environment. Changes in phenotype, genes, and glycosaminoglycan amounts according to each environment were evaluated. Based on this, the mechanism of each culture environment on the intrinsic properties of conserving chondrocytes was suggested. RESULTS Chondrocytes live under pressure from the surrounding collagen tissue and experience a hypoxic environment because collagen inhibits oxygen permeability. By culturing the chondrocytes in a DCP environment, the capability of DCP to produce a low-oxygen and physical pressure environment was verified. When human primary chondrocytes, which require pressure and a low-oxygen environment during culture to maintain their innate properties, were cultured using the hydrogel blanket, the original shapes and properties of the chondrocytes were maintained. The intrinsic properties could be recovered even in aged cells that had lost their original cell properties. CONCLUSIONS A DCP culture method using a biomimetic hydrogel blanket provides cells with an adjustable physical pressure and a low-oxygen environment. Through this technique, we could maintain the original cellular phenotypes and intrinsic properties of human primary chondrocytes. The results of this study can be applied to other cells that require special pressure and oxygen concentration control to maintain their intrinsic properties. Additionally, this technique has the potential to be applied to the re-differentiation of cells that have lost their original properties.
Collapse
Affiliation(s)
- Seung-Hoon Um
- grid.35541.360000000121053345Biomaterials Research Center, Biomedical Research Division, Korea Institute of Science and Technology (KIST), 02792 Seoul, Republic of Korea ,grid.23856.3a0000 0004 1936 8390Laboratory for Biomaterials and Bioengineering, Department of Min-Met-Materials Engineering, Research Center of CHU de Quebec, Division of Regenerative Medicine, Canada Research Chair I in Biomaterials and Bioengineering for the Innovation in Surgery, Laval University, G1V 0A6 Quebec City, Quebec, Canada
| | - Youngmin Seo
- grid.35541.360000000121053345Biomaterials Research Center, Biomedical Research Division, Korea Institute of Science and Technology (KIST), 02792 Seoul, Republic of Korea ,R&D Institute, OID Ltd, Seoul, 06286 Republic of Korea
| | - Hyunseon Seo
- grid.35541.360000000121053345Biomaterials Research Center, Biomedical Research Division, Korea Institute of Science and Technology (KIST), 02792 Seoul, Republic of Korea ,grid.264381.a0000 0001 2181 989XSchool of Medicine, Sungkyunkwan University, Suwon, 16419 Republic of Korea
| | - Kyungwoo Lee
- grid.35541.360000000121053345Biomaterials Research Center, Biomedical Research Division, Korea Institute of Science and Technology (KIST), 02792 Seoul, Republic of Korea
| | - Sun Hwa Park
- grid.23856.3a0000 0004 1936 8390Laboratory for Biomaterials and Bioengineering, Department of Min-Met-Materials Engineering, Research Center of CHU de Quebec, Division of Regenerative Medicine, Canada Research Chair I in Biomaterials and Bioengineering for the Innovation in Surgery, Laval University, G1V 0A6 Quebec City, Quebec, Canada
| | - Jung Ho Jeon
- grid.411947.e0000 0004 0470 4224Department of Otolaryngology-Head and Neck Surgery, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea ,grid.411947.e0000 0004 0470 4224Department of Biomedicine & Health Sciences, Department of Otolaryngology-Head and Neck Surgery, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Jung Yeon Lim
- grid.411947.e0000 0004 0470 4224Department of Otolaryngology-Head and Neck Surgery, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Myoung-Ryul Ok
- grid.35541.360000000121053345Biomaterials Research Center, Biomedical Research Division, Korea Institute of Science and Technology (KIST), 02792 Seoul, Republic of Korea
| | - Yu-Chan Kim
- grid.35541.360000000121053345Biomaterials Research Center, Biomedical Research Division, Korea Institute of Science and Technology (KIST), 02792 Seoul, Republic of Korea ,grid.412786.e0000 0004 1791 8264Division of Bio-Medical Science and Technology, KIST School, Korea University of Science and Technology, Seoul, 02792 Republic of Korea
| | - Hyunjung Kim
- grid.256753.00000 0004 0470 5964Division of Nursing, Research Institute of Nursing Science, Hallym University, Chuncheon, 24252 Republic of Korea
| | - Cheol-Hong Cheon
- grid.222754.40000 0001 0840 2678Department of Chemistry, Korea University, Seoul, 02841 Republic of Korea
| | - Hyung-Seop Han
- grid.35541.360000000121053345Biomaterials Research Center, Biomedical Research Division, Korea Institute of Science and Technology (KIST), 02792 Seoul, Republic of Korea
| | - James R. Edwards
- grid.4991.50000 0004 1936 8948Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences (NDORMS), Botnar Research Centre, University of Oxford, Oxford, OX3 7LD UK
| | - Sung Won Kim
- grid.411947.e0000 0004 0470 4224Department of Otolaryngology-Head and Neck Surgery, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea ,grid.411947.e0000 0004 0470 4224Department of Biomedicine & Health Sciences, Department of Otolaryngology-Head and Neck Surgery, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Hojeong Jeon
- grid.35541.360000000121053345Biomaterials Research Center, Biomedical Research Division, Korea Institute of Science and Technology (KIST), 02792 Seoul, Republic of Korea ,grid.412786.e0000 0004 1791 8264Division of Bio-Medical Science and Technology, KIST School, Korea University of Science and Technology, Seoul, 02792 Republic of Korea ,grid.222754.40000 0001 0840 2678KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841 Republic of Korea
| |
Collapse
|
9
|
Abadi B, Goshtasbi N, Bolourian S, Tahsili J, Adeli-Sardou M, Forootanfar H. Electrospun hybrid nanofibers: Fabrication, characterization, and biomedical applications. Front Bioeng Biotechnol 2022; 10:986975. [PMID: 36561047 PMCID: PMC9764016 DOI: 10.3389/fbioe.2022.986975] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 11/16/2022] [Indexed: 12/05/2022] Open
Abstract
Nanotechnology is one of the most promising technologies available today, holding tremendous potential for biomedical and healthcare applications. In this field, there is an increasing interest in the use of polymeric micro/nanofibers for the construction of biomedical structures. Due to its potential applications in various fields like pharmaceutics and biomedicine, the electrospinning process has gained considerable attention for producing nano-sized fibers. Electrospun nanofiber membranes have been used in drug delivery, controlled drug release, regenerative medicine, tissue engineering, biosensing, stent coating, implants, cosmetics, facial masks, and theranostics. Various natural and synthetic polymers have been successfully electrospun into ultrafine fibers. Although biopolymers demonstrate exciting properties such as good biocompatibility, non-toxicity, and biodegradability, they possess poor mechanical properties. Hybrid nanofibers from bio and synthetic nanofibers combine the characteristics of biopolymers with those of synthetic polymers, such as high mechanical strength and stability. In addition, a variety of functional agents, such as nanoparticles and biomolecules, can be incorporated into nanofibers to create multifunctional hybrid nanofibers. Due to the remarkable properties of hybrid nanofibers, the latest research on the unique properties of hybrid nanofibers is highlighted in this study. Moreover, various established hybrid nanofiber fabrication techniques, especially the electrospinning-based methods, as well as emerging strategies for the characterization of hybrid nanofibers, are summarized. Finally, the development and application of electrospun hybrid nanofibers in biomedical applications are discussed.
Collapse
Affiliation(s)
- Banafshe Abadi
- Herbal and Traditional Medicines Research Center, Kerman University of Medical Sciences, Kerman, Iran,Brain Cancer Research Core (BCRC), Universal Scientific Education and Research Network (USERN), Kerman, Iran
| | - Nazanin Goshtasbi
- Department of Pharmaceutics, Faculty of Pharmacy and Pharmaceutical Sciences, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Saman Bolourian
- Department of Biology, Faculty of Science, Alzahra University, Tehran, Iran
| | - Jaleh Tahsili
- Department of Plant Biology, Faculty of Biological Science, Tarbiat Modares University, Tehran, Iran
| | - Mahboubeh Adeli-Sardou
- Medical Mycology and Bacteriology Research Center, Kerman University of Medical Sciences, Kerman, Iran,Student Research Committee, Kerman University of Medical Sciences, Kerman, Iran,*Correspondence: Mahboubeh Adeli-Sardou, ; Hamid Forootanfar,
| | - Hamid Forootanfar
- Pharmaceutical Sciences and Cosmetic Products Research Center, Kerman University of Medical Sciences, Kerman, Iran,Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Kerman University of Medical Sciences, Kerman, Iran,*Correspondence: Mahboubeh Adeli-Sardou, ; Hamid Forootanfar,
| |
Collapse
|
10
|
Bakhtiary N, Pezeshki-Modaress M, Najmoddin N. Wet-electrospinning of nanofibrous magnetic composite 3-D scaffolds for enhanced stem cells neural differentiation. Chem Eng Sci 2022. [DOI: 10.1016/j.ces.2022.118144] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
11
|
Kim W, Park E, Yoo HS, Park J, Jung YM, Park JH. Recent Advances in Monitoring Stem Cell Status and Differentiation Using Nano-Biosensing Technologies. NANOMATERIALS (BASEL, SWITZERLAND) 2022; 12:2934. [PMID: 36079970 PMCID: PMC9457759 DOI: 10.3390/nano12172934] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 08/22/2022] [Accepted: 08/23/2022] [Indexed: 05/14/2023]
Abstract
In regenerative medicine, cell therapies using various stem cells have received attention as an alternative to overcome the limitations of existing therapeutic methods. Clinical applications of stem cells require the identification of characteristics at the single-cell level and continuous monitoring during expansion and differentiation. In this review, we recapitulate the application of various stem cells used in regenerative medicine and the latest technological advances in monitoring the differentiation process of stem cells. Single-cell RNA sequencing capable of profiling the expression of many genes at the single-cell level provides a new opportunity to analyze stem cell heterogeneity and to specify molecular markers related to the branching of differentiation lineages. However, this method is destructive and distorted. In addition, the differentiation process of a particular cell cannot be continuously tracked. Therefore, several spectroscopic methods have been developed to overcome these limitations. In particular, the application of Raman spectroscopy to measure the intrinsic vibration spectrum of molecules has been proposed as a powerful method that enables continuous monitoring of biochemical changes in the process of the differentiation of stem cells. This review provides a comprehensive overview of current analytical methods employed for stem cell engineering and future perspectives of nano-biosensing technologies as a platform for the in situ monitoring of stem cell status and differentiation.
Collapse
Affiliation(s)
- Wijin Kim
- Department of Biomedical Science, Kangwon National University, Chuncheon 24341, Gangwon-do, Korea
| | - Eungyeong Park
- Department of Chemistry, Kangwon National University, Chuncheon 24341, Gangwon-do, Korea
| | - Hyuk Sang Yoo
- Department of Biomedical Science, Kangwon National University, Chuncheon 24341, Gangwon-do, Korea
- Kangwon Radiation Convergence Research Support Center, Kangwon National University, Chuncheon 24341, Gangwon-do, Korea
| | - Jongmin Park
- Department of Chemistry, Kangwon National University, Chuncheon 24341, Gangwon-do, Korea
| | - Young Mee Jung
- Department of Chemistry, Kangwon National University, Chuncheon 24341, Gangwon-do, Korea
- Kangwon Radiation Convergence Research Support Center, Kangwon National University, Chuncheon 24341, Gangwon-do, Korea
| | - Ju Hyun Park
- Department of Biomedical Science, Kangwon National University, Chuncheon 24341, Gangwon-do, Korea
| |
Collapse
|
12
|
Tambrchi P, Mahdavi AH, DaliriJoupari M, Soltani L. Polycaprolactone-co-polylactic acid nanofiber scaffold in combination with 5-azacytidine and transforming growth factor-β to induce cardiomyocyte differentiation of adipose-derived mesenchymal stem cells. Cell Biochem Funct 2022; 40:668-682. [PMID: 35924670 DOI: 10.1002/cbf.3728] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 06/10/2022] [Accepted: 06/13/2022] [Indexed: 11/11/2022]
Abstract
Adipose-derived mesenchymal stem cells (Ad-MSCs) are promising candidates for cardiac repair/regeneration. The application of copolymer nanoscaffolds has received great attention in tissue engineering to support differentiation and functional tissue organization toward effective tissue regeneration. The objective of the current study was to develop functional and bioactive scaffolds by combining polycaprolactone (PCL) and polylactic acid (PLA) for cardiomyocyte differentiation of human Ad-MSC (hAd-MSCs) in the absence or presence of 5-azacytidine and transforming growth factor-β (TGF-β). To that end, the human MSCs were extracted from human adipose tissue (AD). The cardiomyocyte differentiation potency of hAd-MSCs was evaluated on the novel synthetic PCL/PLA nanofiber scaffolds prepared in the absence and presence of 5-azacytidine and TGF-β supplements. A PCL/PLA nanofibrous scaffold was fabricated using the electrospinning method and its nanotopography and porous structure were characterized using scanning electron microscopy. In addition, the attachment of hAd-MSCs on the PCL/PLA scaffolds was semiquantitatively investigated. Compared with other treatments, the PCL/PLA nanofibrous scaffold supplemented with both 5-azacytidine and TGF-β was observed to differentiate hAd-MSCs into cardiomyocytes at Day 21 as evidenced by real-time PCR for cardiac-specific genes including cardiac troponin I (cTnI), GATA4, MYH7, and NKX2.5. In addition, flow cytometric analysis of cTnI-positive cells demonstrated that the cardiomyocyte differentiation of hAd-MSCs was more efficient on the PCL/PLA nanofibrous scaffold supplemented with both 5-azacytidine and TGF-β than it was in the other treatment groups. Generally speaking, the results show that PCL/PLA nanofibrous scaffolds may be applied as a platform for efficient differentiation of hAd-MSCs into functional cardiomyocytes.
Collapse
Affiliation(s)
- Parastoo Tambrchi
- Department of Animal Biotechnology, National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran
| | - Amir Hossein Mahdavi
- Department of Animal Science, College of Agriculture, Isfahan University of Technology, Isfahan, Iran
| | - Morteza DaliriJoupari
- Department of Animal Biotechnology, National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran
| | - Leila Soltani
- Department of Animal Sciences, College of Agriculture and Natural Resources, Razi University, Kermanshah, Iran
| |
Collapse
|
13
|
Sarviya N, Basu SM, Mani R, Chauhan M, Kingshott P, Giri J. Biomimicking nanofibrous gelatin microspheres recreating the stem cell niche for their ex-vivo expansion and in-vivo like differentiation for injectable stem cell transplantation. BIOMATERIALS ADVANCES 2022; 139:212981. [PMID: 35882137 DOI: 10.1016/j.bioadv.2022.212981] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 06/02/2022] [Accepted: 06/08/2022] [Indexed: 06/15/2023]
Abstract
Stem cells based novel treatment modality for degenerative and immune dysfunction diseases created a huge demand of suitable carriers to support ex-vivo production of quality stem cells, and effective in-vivo transplantation of stem cells and their fate. In spite of promising candidature of nanofibrous microspheres (NFM) to recreate native stem cell niches to be used for possible scaling-up for ex-vivo stem cells expansion, it remains fairly unexplored. A systematic study on the stem cell-NFM interaction comparative with commercial microspheres (CM) has been performed for the first time. Gelatin NFM with variable physicochemical properties such as size, surface properties, surface chemistry, and variable degradability were prepared using microemulsion coupled with thermally induced phase separation (TIPS) method. Effect of physicochemical properties of NFM and their cellular interaction such as binding, morphology, metabolic activity and proliferation studies were performed using human bone marrow-derived mesenchymal stem cells (hBMSCs), human dental follicle stem cells (hDFSCs) and human gingival fibroblast (HGF) cells and compared with the commercial and solid microspheres. Gelatin NFM supports excellent cell binding, proliferation, metabolic activities and chemical cues specific differentiation. All out-turns indicate that NFM stand to be an outstanding candidate for ex-vivo cells' expansion and injectable carriers for stem cell transplantation.
Collapse
Affiliation(s)
- Nandini Sarviya
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Telangana, India; Department of Chemistry and Biotechnology, Swinburne Institute of Technology, Victoria, Australia
| | - Suparna Mercy Basu
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Telangana, India
| | - Rajesh Mani
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Telangana, India
| | - Meenakshi Chauhan
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Telangana, India
| | - Peter Kingshott
- Department of Chemistry and Biotechnology, Swinburne Institute of Technology, Victoria, Australia
| | - Jyotsnendu Giri
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Telangana, India.
| |
Collapse
|
14
|
Shirehjini LM, Sharifi F, Shojaei S, Irani S. Poly-caprolactone nanofibrous coated with sol-gel alginate/ mesenchymal stem cells for cartilage tissue engineering. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
15
|
Liu X, Liu Y, He H, Xiang W, He C. Human adipose and synovial mesenchymal stem cells improve osteoarthritis in rats by reducing chondrocyte reactive oxygen species and inhibiting inflammatory response. J Clin Lab Anal 2022; 36:e24353. [PMID: 35312120 PMCID: PMC9102617 DOI: 10.1002/jcla.24353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 03/02/2022] [Accepted: 03/08/2022] [Indexed: 12/01/2022] Open
Abstract
Background We explored the therapeutic effects of Adipose‐derived mesenchymal stem cells (ADMSCs) and Synovial‐derived mesenchymal stem cells (SDMSCs) on osteoarthritis (OA). Methods SDMSCs and ADMSCs were co‐cultured with chondrocytes and stimulated with interleukin (IL)‐1β. An OA model was established on rats by intra‐articular injection with ADMSCs and SDMSCs. After 8 weeks, the joint diameter difference was detected, and histological staining was used to observe the pathological changes in cartilage tissue. Enzyme‐linked immunosorbent assay (ELISA) was used to detect the expressions of IL‐6, tumor necrosis factor (TNF)‐α and IL‐1β in joint fluid. The expressions of COL2A1, Aggrecan, Matrix metalloproteinase (MMP)‐13, SOX9, IL‐6, TNF‐α and IL‐1β were detected by qRT‐PCR and Western blotting in cartilage tissue. Reactive oxygen species (ROS) content in cells and cartilage tissues was detected by ROS kit. Results SDMSCs and ADMSCs co‐cultured with chondrocytes could reduce MMP‐13 expression, increase the expressions of COL2A1, Aggrecan and SOX9, as well as reverse the effects of IL‐1β on promoting ROS content and inflammatory factors levels. After the OA model was established, the injection of ADMSCs and SDMSCs reduced the differences in joint diameter and tissue lesions in OA rats. The OA model led to increased levels of IL‐6, TNF‐α and IL‐1β in joint fluid and cartilage tissue, while the injection of ADMSCs and SDMSCs inhibited the inflammatory factor levels in OA rats, and increased the expressions of COL2A1, Aggrecan and SOX9 in OA rats. Conclusion ADMSCs and SDMSCs improve osteoarthritis in rats by reducing chondrocyte ROS and inhibiting inflammatory response.
Collapse
Affiliation(s)
- Xunzhi Liu
- Orthopedics Department First Affiliated Hospital of Gannan Medical University Ganzhou City China
| | - Yaqing Liu
- Pediatric Department First Affiliated Hospital of Gannan Medical University Ganzhou City China
| | - Huabin He
- Orthopedics Department First Affiliated Hospital of Gannan Medical University Ganzhou City China
| | - Weiwei Xiang
- Orthopedics Department First Affiliated Hospital of Gannan Medical University Ganzhou City China
| | - Cheng He
- Orthopedics Department First Affiliated Hospital of Gannan Medical University Ganzhou City China
| |
Collapse
|
16
|
Akbari A, Rabbani S, Irani S, Zandi M, Sharifi F, Ameli F, Mohamadali M. In vitro and in vivo study of carboxymethyl chitosan/polyvinyl alcohol for wound dressing application. J Appl Polym Sci 2022. [DOI: 10.1002/app.51764] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Alireza Akbari
- Department of Biology, Science and Research Branch Islamic Azad University Tehran Iran
| | - Shahram Rabbani
- Tehran Heart Center Tehran University of Medical Sciences Tehran Iran
| | - Shiva Irani
- Department of Biology, Science and Research Branch Islamic Azad University Tehran Iran
| | - Mojgan Zandi
- Department of Biomaterial Iran Polymer and Petrochemical Institute Tehran Iran
| | - Fereshteh Sharifi
- Hard Tissue Engineering Research Center Tissue Engineering and Regenerative Medicine Institute, Central Tehran Branch, Islamic Azad University Tehran Iran
| | - Fereshteh Ameli
- Department of Pathology Cancer Institute, Imam Khomeini Hospital Complex, Tehran University of Medical Science Tehran Iran
| | - Marjan Mohamadali
- Department of Biology, Science and Research Branch Islamic Azad University Tehran Iran
| |
Collapse
|
17
|
Applying extrusion-based 3D printing technique accelerates fabricating complex biphasic calcium phosphate-based scaffolds for bone tissue regeneration. J Adv Res 2021; 40:69-94. [PMID: 36100335 PMCID: PMC9481949 DOI: 10.1016/j.jare.2021.12.012] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 12/09/2021] [Accepted: 12/23/2021] [Indexed: 12/17/2022] Open
Abstract
Biphasic calcium phosphates offer a chemically similar biomaterial to the natural bone, which can significantly accelerate bone formation and reconstruction. Robocasting is a suitable technique to produce porous scaffolds supporting cell viability, proliferation, and differentiation. This review discusses materials and methods utilized for BCP robocasting, considering recent advancements and existing challenges in using additives for bioink preparation. Commercialization and marketing approach, in-vitro and in-vivo evaluations, biologic responses, and post-processing steps are also investigated. Possible strategies and opportunities for the use of BCP toward injured bone regeneration along with clinical applications are discussed. The study proposes that BCP possesses an acceptable level of bone substituting, considering its challenges and struggles.
Background Aim of review Key scientific concepts of review
Collapse
|
18
|
Fazaeli H, Kalhor N, Naserpour L, Davoodi F, Sheykhhasan M, Hosseini SKE, Rabiei M, Sheikholeslami A. A Comparative Study on the Effect of Exosomes Secreted by Mesenchymal Stem Cells Derived from Adipose and Bone Marrow Tissues in the Treatment of Osteoarthritis-Induced Mouse Model. BIOMED RESEARCH INTERNATIONAL 2021; 2021:9688138. [PMID: 34616850 PMCID: PMC8490078 DOI: 10.1155/2021/9688138] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 08/25/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND Exosomes as extracellular vesicles (EVs) are nanoscale intercellular messengers secreted from cells to deliver biological signals. Today, exosomes have become a new field of research in regenerative medicine and are considered as potential therapies to control inflammation and wound healing and enhance and improve healing in many diseases. Given the global burden of osteoarthritis (OA) as the fastest-growing health condition and one of the major causes of physical disability in the aging population, research to establish EVs as therapeutic products can meet the basic clinical needs in the management of osteoarthritis and provide a therapeutic solution. OBJECTIVES The present study is aimed at evaluating the regenerative potentials of the exosomes secreted from adipose and bone marrow tissue-derived mesenchymal stem cells (AD- and BM-MSCs) in ameliorating the symptoms of OA. METHOD In this experimental study, AD- and BM-MSCs were isolated and cultured in the laboratory until passage 3. Finally, these cells' secreted exosomes were isolated from their conditioned medium. Ciprofloxacin-induced OA mouse models underwent intra-articular injection of exosomes from AD-MSCs and BM-MSCs. Finally, the expression levels of collagen I and II, sox9, and aggrecan genes using real-time PCR, histological analysis, and immunohistochemical (IHC) studies were performed. RESULTS Real-time PCR data showed that although the expression level of collagen type II was lower in both exosome-treated groups than the normal, but it was significantly increased in comparison with the sham and OA, with higher expression in BM-Exo rather than AD-Exo group. Similarly, the histological staining and IHC results have provided almost identical data, emphasizing on better therapeutic effect of BM-MSCs-exosome than AD-MSCs-exosome. CONCLUSION BM-MSCs secreted exosomes in comparison with AD-MSCs could be considered as a better therapeutic option to improve osteoarthritis and exhibit potential as a disease-modifying osteoarthritis cell-free product.
Collapse
Affiliation(s)
- Hoda Fazaeli
- Department of Mesenchymal Stem Cells, Academic Center for Education, Culture, and Research (ACECR), Qom Branch, Qom, Iran
| | - Naser Kalhor
- Department of Mesenchymal Stem Cells, Academic Center for Education, Culture, and Research (ACECR), Qom Branch, Qom, Iran
| | - Leila Naserpour
- Department of Reproductive Biology, Academic Center for Education, Culture, and Research (ACECR), Qom Branch, Qom, Iran
| | - Faezeh Davoodi
- Department of Mesenchymal Stem Cells, Academic Center for Education, Culture, and Research (ACECR), Qom Branch, Qom, Iran
| | - Mohsen Sheykhhasan
- Department of Mesenchymal Stem Cells, Academic Center for Education, Culture, and Research (ACECR), Qom Branch, Qom, Iran
| | | | - Mohammad Rabiei
- Department of Biology, Faculty of Science, Azad Islamic University of Qom, Qom, Iran
| | - Azar Sheikholeslami
- Department of Mesenchymal Stem Cells, Academic Center for Education, Culture, and Research (ACECR), Qom Branch, Qom, Iran
| |
Collapse
|
19
|
Merimi M, El-Majzoub R, Lagneaux L, Moussa Agha D, Bouhtit F, Meuleman N, Fahmi H, Lewalle P, Fayyad-Kazan M, Najar M. The Therapeutic Potential of Mesenchymal Stromal Cells for Regenerative Medicine: Current Knowledge and Future Understandings. Front Cell Dev Biol 2021; 9:661532. [PMID: 34490235 PMCID: PMC8416483 DOI: 10.3389/fcell.2021.661532] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Accepted: 05/28/2021] [Indexed: 12/11/2022] Open
Abstract
In recent decades, research on the therapeutic potential of progenitor cells has advanced considerably. Among progenitor cells, mesenchymal stromal cells (MSCs) have attracted significant interest and have proven to be a promising tool for regenerative medicine. MSCs are isolated from various anatomical sites, including bone marrow, adipose tissue, and umbilical cord. Advances in separation, culture, and expansion techniques for MSCs have enabled their large-scale therapeutic application. This progress accompanied by the rapid improvement of transplantation practices has enhanced the utilization of MSCs in regenerative medicine. During tissue healing, MSCs may exhibit several therapeutic functions to support the repair and regeneration of injured tissue. The process underlying these effects likely involves the migration and homing of MSCs, as well as their immunotropic functions. The direct differentiation of MSCs as a cell replacement therapeutic mechanism is discussed. The fate and behavior of MSCs are further regulated by their microenvironment, which may consequently influence their repair potential. A paracrine pathway based on the release of different messengers, including regulatory factors, chemokines, cytokines, growth factors, and nucleic acids that can be secreted or packaged into extracellular vesicles, is also implicated in the therapeutic properties of MSCs. In this review, we will discuss relevant outcomes regarding the properties and roles of MSCs during tissue repair and regeneration. We will critically examine the influence of the local microenvironment, especially immunological and inflammatory signals, as well as the mechanisms underlying these therapeutic effects. Importantly, we will describe the interactions of local progenitor and immune cells with MSCs and their modulation during tissue injury. We will also highlight the crucial role of paracrine pathways, including the role of extracellular vesicles, in this healing process. Moreover, we will discuss the therapeutic potential of MSCs and MSC-derived extracellular vesicles in the treatment of COVID-19 (coronavirus disease 2019) patients. Overall, this review will provide a better understanding of MSC-based therapies as a novel immunoregenerative strategy.
Collapse
Affiliation(s)
- Makram Merimi
- Laboratory of Experimental Hematology, Institut Jules Bordet, Université Libre de Bruxelles (ULB), Bruxelles, Belgium.,LBBES Laboratory, Genetics and Immune-Cell Therapy Unit, Faculty of Sciences, University Mohammed Premier, Oujda, Morocco
| | - Rania El-Majzoub
- Department of Biomedical Sciences, School of Pharmacy, Lebanese International University, Beirut, Lebanon.,Laboratory of Cancer Biology and Molecular Immunology, Faculty of Sciences-I, Lebanese University, Beirut, Lebanon
| | - Laurence Lagneaux
- Laboratory of Clinical Cell Therapy, Institut Jules Bordet, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Douâa Moussa Agha
- Laboratory of Experimental Hematology, Institut Jules Bordet, Université Libre de Bruxelles (ULB), Bruxelles, Belgium
| | - Fatima Bouhtit
- Laboratory of Experimental Hematology, Institut Jules Bordet, Université Libre de Bruxelles (ULB), Bruxelles, Belgium.,LBBES Laboratory, Genetics and Immune-Cell Therapy Unit, Faculty of Sciences, University Mohammed Premier, Oujda, Morocco
| | - Nathalie Meuleman
- Laboratory of Experimental Hematology, Institut Jules Bordet, Université Libre de Bruxelles (ULB), Bruxelles, Belgium
| | - Hassan Fahmi
- Osteoarthritis Research Unit, University of Montreal Hospital Research Center (CRCHUM), Montreal, QC, Canada
| | - Philippe Lewalle
- Laboratory of Experimental Hematology, Institut Jules Bordet, Université Libre de Bruxelles (ULB), Bruxelles, Belgium
| | - Mohammad Fayyad-Kazan
- Laboratory of Cancer Biology and Molecular Immunology, Faculty of Sciences-I, Lebanese University, Beirut, Lebanon.,Department of Natural Sciences, School of Arts and Sciences, Lebanese American University, Beirut, Lebanon
| | - Mehdi Najar
- Laboratory of Clinical Cell Therapy, Institut Jules Bordet, Université Libre de Bruxelles (ULB), Brussels, Belgium.,Osteoarthritis Research Unit, University of Montreal Hospital Research Center (CRCHUM), Montreal, QC, Canada
| |
Collapse
|
20
|
Lee SJ, Nam Y, Rim YA, Lee K, Ju JH, Kim DS. Perichondrium-inspired permeable nanofibrous tube well promoting differentiation of hiPSC-derived pellet toward hyaline-like cartilage pellet. Biofabrication 2021; 13. [PMID: 34404032 DOI: 10.1088/1758-5090/ac1e76] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 08/17/2021] [Indexed: 01/22/2023]
Abstract
The pellet formation has been regarded as a golden standard forin vitrochondrogenic differentiation. However, a spatially inhomogeneous chondrogenic microenvironment around a pellet resulted from the use of a traditional impermeable narrow tube, such as the conical tube, undermines the differentiation performance and therapeutic potential of differentiated cartilage pellet in defective articular cartilage treatment. To address this drawback, a perichondrium-inspired permeable nanofibrous tube (PINaT) well with a nanofibrous wall permeable to gas and soluble molecules is proposed. The PINaT well was fabricated with a micro deep drawing process where a flat thin nanofibrous membrane was transformed to a 3.5 mm deep tube well with a ∼50µm thick nanofibrous wall. Similar toin vivoperichondrium, the PINaT well was found to allow oxygen and growth factor diffusion required for chondrogenic differentiation across the entire nanofibrous wall. Analyses of gene expressions (COL2A1, COL10A1, ACAN, and SOX9), proteins (type II and X collagen), and glycosaminoglycans contents were conducted to assess the differentiation performance and clinical efficacy of differentiated cartilage pellet. The regulated spatially homogeneous chondrogenic microenvironment around the human induced pluripotent stem cell-derived pellet (3 × 105cells per pellet) in the PINaT well remarkably improved the quality of the differentiated pellet toward a more hyaline-like cartilage pellet. Furthermore, an accelerated chondrogenic differentiation process of the pellet produced by the PINaT well was achieved for 14 days, demonstrating a hyaline cartilage-specific marker similar to the control pellet differentiated for 20 days. Finally, the enhanced clinical efficacy of the hyaline-like cartilage pellet was confirmed using an osteochondral defect rat model, with the repaired tissue resembling hyaline cartilage rather than fibrous cartilage after 8 weeks of regeneration.
Collapse
Affiliation(s)
- Seong Jin Lee
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), 77, Cheongam-ro, Nam-gu, Pohang, Gyeongbuk 37673, Republic of Korea
| | - Yoojun Nam
- Catholic iPSC Research Center, College of Medicine, The Catholic University of Korea, 20 Banpo-daero, Seocho-gu, Seoul 06591, Republic of Korea.,Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary's Hospital, Institute of Medical Science, College of Medicine, The Catholic University of Korea, 20 Banpo-daero, Seocho-gu, Seoul 06591, Republic of Korea
| | - Yeri Alice Rim
- Catholic iPSC Research Center, College of Medicine, The Catholic University of Korea, 20 Banpo-daero, Seocho-gu, Seoul 06591, Republic of Korea.,Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary's Hospital, Institute of Medical Science, College of Medicine, The Catholic University of Korea, 20 Banpo-daero, Seocho-gu, Seoul 06591, Republic of Korea
| | - Kijun Lee
- Catholic iPSC Research Center, College of Medicine, The Catholic University of Korea, 20 Banpo-daero, Seocho-gu, Seoul 06591, Republic of Korea
| | - Ji Hyeon Ju
- Catholic iPSC Research Center, College of Medicine, The Catholic University of Korea, 20 Banpo-daero, Seocho-gu, Seoul 06591, Republic of Korea.,Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary's Hospital, Institute of Medical Science, College of Medicine, The Catholic University of Korea, 20 Banpo-daero, Seocho-gu, Seoul 06591, Republic of Korea
| | - Dong Sung Kim
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), 77, Cheongam-ro, Nam-gu, Pohang, Gyeongbuk 37673, Republic of Korea.,Department of Chemical Engineering, Pohang University of Science and Technology (POSTECH), 77, Cheongam-ro, Nam-gu, Pohang, Gyeongbuk 37673, Republic of Korea.,Institute for Convergence Research and Education in Advanced Technology, Yonsei University, 50, Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| |
Collapse
|
21
|
Fülber J, Agreste FR, Seidel SRT, Sotelo EDP, Barbosa ÂP, Michelacci YM, Baccarin RYA. Chondrogenic potential of mesenchymal stem cells from horses using a magnetic 3D cell culture system. World J Stem Cells 2021; 13:645-658. [PMID: 34249233 PMCID: PMC8246251 DOI: 10.4252/wjsc.v13.i6.645] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 03/29/2021] [Accepted: 06/04/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Mesenchymal stem cells (MSCs) represent a promising therapy for the treatment of equine joint diseases, studied due to their possible immunomodulatory characteristics and regenerative capacity. However, the source of most suitable MSCs for producing cartilage for regenerative processes in conjunction with biomaterials for an enhanced function is yet to be established. AIM To compare the chondrogenicity of MSCs derived from synovial fluid, bone marrow, and adipose tissue of horses, using the aggrecan synthesis. METHODS MSCs from ten horses were cultured, phenotypic characterization was done with antibodies CD90, CD44 and CD34 and were differentiated into chondrocytes. The 3D cell culture system in which biocompatible nanoparticles consisting of gold, iron oxide, and poly-L-lysine were added to the cells, and they were forced by magnets to form one microspheroid. The microspheroids were exposed to a commercial culture medium for 4 d, 7 d, 14 d, and 21 d. Proteoglycan extraction was performed, and aggrecan was quantified by enzyme-linked immunosorbent assay. Keratan sulfate and aggrecan in the microspheroids were identified and localized by immunofluorescence. RESULTS All cultured cells showed fibroblast-like appearance, the ability to adhere to the plastic surface, and were positive for CD44 and CD90, thus confirming the characteristics and morphology of MSCs. The soluble protein concentrations were higher in the microspheroids derived from adipose tissue. The aggrecan concentration and the ratio of aggrecan to soluble proteins were higher in microspheroids derived from synovial fluid than in those derived from bone marrow, thereby showing chondrogenic superiority. Microspheroids from all sources expressed aggrecan and keratan sulfate when observed using confocal immunofluorescence microscopy. All sources of MSCs can synthesize aggrecan, however, MSCs from synovial fluid and adipose tissue have demonstrated better biocompatibility in a 3D environment, thus suggesting chondrogenic superiority. CONCLUSION All sources of MSCs produce hyaline cartilage; however, the use of synovial liquid or adipose tissue should be recommended when it is intended for use with biomaterials or scaffolds.
Collapse
Affiliation(s)
- Joice Fülber
- Departamento de Clínica Médica, Medicina Veterinária e Zootecnia, Universidade de São Paulo, São Paulo 05506-270, Brazil.
| | - Fernanda R Agreste
- Departamento de Clínica Médica, Medicina Veterinária e Zootecnia, Universidade de São Paulo, São Paulo 05506-270, Brazil
| | - Sarah R T Seidel
- Departamento de Clínica Médica, Medicina Veterinária e Zootecnia, Universidade de São Paulo, São Paulo 05506-270, Brazil
| | - Eric D P Sotelo
- Departamento de Clínica Médica, Medicina Veterinária e Zootecnia, Universidade de São Paulo, São Paulo 05506-270, Brazil
| | - Ângela P Barbosa
- Departamento de Clínica Médica, Medicina Veterinária e Zootecnia, Universidade de São Paulo, São Paulo 05506-270, Brazil
| | - Yara M Michelacci
- Departamento de Bioquímica, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo 04044-020, Brazil
| | - Raquel Y A Baccarin
- Departamento de Clínica Médica, Medicina Veterinária e Zootecnia, Universidade de São Paulo, São Paulo 05506-270, Brazil
| |
Collapse
|
22
|
Karimi S, Bagher Z, Najmoddin N, Simorgh S, Pezeshki-Modaress M. Alginate-magnetic short nanofibers 3D composite hydrogel enhances the encapsulated human olfactory mucosa stem cells bioactivity for potential nerve regeneration application. Int J Biol Macromol 2020; 167:796-806. [PMID: 33278440 DOI: 10.1016/j.ijbiomac.2020.11.199] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 11/22/2020] [Accepted: 11/29/2020] [Indexed: 12/18/2022]
Abstract
The design of 3D hydrogel constructs to elicit highly controlled cell response is a major field of interest in developing tissue engineering. The bioactivity of encapsulated cells inside pure alginate hydrogel is limited by its relatively inertness. Combining short nanofibers within a hydrogel serves as a promising method to develop a cell friendly environment mimicking the extracellular matrix. In this paper, we fabricated alginate hydrogels incorporating different magnetic short nanofibers (M.SNFs) content for olfactory ecto-mesenchymal stem cells (OE-MSCs) encapsulation. Wet-electrospun gelatin and superparamagnetic iron oxide nanoparticles (SPIONs) nanocomposite nanofibers were chopped using sonication under optimized conditions and subsequently embedded in alginate hydrogels. The storage modulus of hydrogel without M.SNFs as well as with 1 and 5 mg/mL of M.SNFs were in the range of nerve tissue. For cell encapsulation, OE-MSCs were used as a new hope for neuronal regeneration due to their neural crest origin. Resazurin analyses and LIVE/DEAD staining confirmed that the composite hydrogels containing M.SNFs can preserve the cell viability after 7 days. Moreover, the proliferation rate was enhanced in M.SNF/hydrogels compared to alginate hydrogel. The presence of SPIONs in the short nanofibers can accelerate neural-like differentiation of OE-MSCs rather than the sample without SPIONs.
Collapse
Affiliation(s)
- Sarah Karimi
- Department of Biomedical Engineering, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Zohreh Bagher
- ENT and Head & Neck Research Center and Department, The Five Senses Institute, Hazrat Rasoul Akram Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Najmeh Najmoddin
- Department of Biomedical Engineering, Science and Research Branch, Islamic Azad University, Tehran, Iran.
| | - Sara Simorgh
- Department of Tissue Engineering and Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | | |
Collapse
|
23
|
Gottschalk J, Elling L. Current state on the enzymatic synthesis of glycosaminoglycans. Curr Opin Chem Biol 2020; 61:71-80. [PMID: 33271474 DOI: 10.1016/j.cbpa.2020.09.008] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 09/23/2020] [Accepted: 09/28/2020] [Indexed: 12/17/2022]
Abstract
Glycosaminoglycans (GAGs) are linear anionic polysaccharides, and most of them show a specific sulfation pattern. GAGs have been studied for decades, and still, new biological functions are discovered. Hyaluronic acid and heparin are sold for medical or cosmetic applications. With increased market and applications, the production of GAGs stays in the focus of research groups and the industry. Common industrial GAG production relies on the extraction of animal tissue. Contamination, high dispersity, and uncontrolled sulfation pattern are still obstacles to this process. Tailored production strategies for the chemoenzymatic synthesis have been developed to address these obstacles. In recent years, enzyme cascades, including uridine-5'-diphosphate sugar syntheses, were established to obtain defined polymer size and dispersity, as well as defined sulfation patterns. Nevertheless, the complex synthesis of GAGs is still a challenging research field.
Collapse
Affiliation(s)
- Johannes Gottschalk
- Laboratory for Biomaterials, Institute of Biotechnology and Helmholtz-Institute for Biomedical Engineering, RWTH Aachen University, Pauwelsstraße 20, 52074, Aachen, Germany
| | - Lothar Elling
- Laboratory for Biomaterials, Institute of Biotechnology and Helmholtz-Institute for Biomedical Engineering, RWTH Aachen University, Pauwelsstraße 20, 52074, Aachen, Germany.
| |
Collapse
|
24
|
Enzymatic Synthesis of Glycans and Glycoconjugates. ADVANCES IN BIOCHEMICAL ENGINEERING/BIOTECHNOLOGY 2020; 175:231-280. [PMID: 33052414 DOI: 10.1007/10_2020_148] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Glycoconjugates have great potential to improve human health in a multitude of different ways and fields. Prominent examples are human milk oligosaccharides and glycosaminoglycans. The typical choice for the production of homogeneous glycoconjugates is enzymatic synthesis. Through the availability of expression and purification protocols, recombinant Leloir glycosyltransferases are widely applied as catalysts for the synthesis of a wide range of glycoconjugates. Extensive utilization of these enzymes also depends on the availability of activated sugars as building blocks. Multi-enzyme cascades have proven a versatile technique to synthesize and in situ regenerate nucleotide sugar.In this chapter, the functions and mechanisms of Leloir glycosyltransferases are revisited, and the advantage of prokaryotic sources and production systems is discussed. Moreover, in vivo and in vitro pathways for the synthesis of nucleotide sugar are reviewed. In the second part, recent and prominent examples of the application of Leloir glycosyltransferase are given, i.e., the synthesis of glycosaminoglycans, glycoconjugate vaccines, and human milk oligosaccharides as well as the re-glycosylation of biopharmaceuticals, and the status of automated glycan assembly is revisited.
Collapse
|
25
|
Amniotic fluid mesenchymal stromal cells from early stages of embryonic development have higher self-renewal potential. In Vitro Cell Dev Biol Anim 2020; 56:701-714. [PMID: 33029689 DOI: 10.1007/s11626-020-00511-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 09/11/2020] [Indexed: 12/21/2022]
Abstract
Amniotic fluid (AF) is a rich source of mesenchymal stromal cells (MSCs) that have the ability to differentiate into multiple lineages rendering them a promising and powerful tool for regenerative medicine. However, information regarding the differences among AFMSCs derived from different gestational stages is limited. In the present study, AFMSCs derived from 125 pregnant rats at four embryonic day (E) stages (E12, E15, E18, and E21) were isolated and cultured. The primary E15 cells were the smallest in size and the easiest to culture and usually grew in a spherical shape that resembled the growth morphology of embryonic stem cells (ESCs). Once adhered, the E12 and E15 AFMSCs grew faster and could be passaged more than 60 times while still maintaining a continuous proliferative state; however, AFMSCs derived from E18 and E21 could normally be maintained for only 10 passages. To identify the possible reasons for this difference, RT-qPCR was used to examine several genes associated with self-renewal ability and cell origin. The Sox2 expression levels indicated that AFMSCs from E12 and E15 possessed stronger self-renewal capability. The K19, Col2A1, FGF5, AFP, and SPC expression levels indicated there were mixed-population cells co-existing in the AFMSC culture. In conclusion, E15 cells were easier to culture than E12 cells, could be passaged more often, and had a higher Sox2 expression than E18 or E21 cells. The E15-derived AFMSCs had higher viability and proliferative capacity than cells from the later stages. Therefore, AF cells from the early stages could be a good choice for exploring potential treatments involving AFMSCs.
Collapse
|
26
|
Irani S, Tavakkoli S, Pezeshki‐Modaress M, Taghavifar E, Mohammadali M, Daemi H. Electrospun nanofibrous alginate sulfate scaffolds promote mesenchymal stem cells differentiation to chondrocytes. J Appl Polym Sci 2020. [DOI: 10.1002/app.49868] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Shiva Irani
- Department of Biology, Science and Research Branch Islamic Azad University Tehran Iran
| | - Sajjad Tavakkoli
- Department of Biology, Science and Research Branch Islamic Azad University Tehran Iran
| | | | - Elham Taghavifar
- Department of Biology, Science and Research Branch Islamic Azad University Tehran Iran
| | - Marjan Mohammadali
- Department of Biology, Science and Research Branch Islamic Azad University Tehran Iran
| | - Hamed Daemi
- Department of Cell Engineering Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR Tehran Iran
| |
Collapse
|
27
|
Han Y, Lian M, Sun B, Jia B, Wu Q, Qiao Z, Dai K. Preparation of high precision multilayer scaffolds based on Melt Electro-Writing to repair cartilage injury. Theranostics 2020; 10:10214-10230. [PMID: 32929344 PMCID: PMC7481411 DOI: 10.7150/thno.47909] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 08/03/2020] [Indexed: 12/22/2022] Open
Abstract
Rationale: Articular cartilage injury is quite common. However, post-injury cartilage repair is challenging and often requires medical intervention, which can be aided by 3D printed tissue engineering scaffolds. Specifically, the high accuracy of Melt Electro-Writing (MEW) technology facilitates the printing of scaffolds that imitate the structure and composition of natural cartilage to promote repair. Methods: MEW and Inkjet printing technology was employed to manufacture a composite scaffold that was then implanted into a cartilage injury site through microfracture surgery. While printing polycaprolactone (PCL) or PCL/hydroxyapatite (HA) scaffolds, cytokine-containing microspheres were sprayed alternately to form multiple layers containing transforming growth factor-β1 and bone morphogenetic protein-7 (surface layer), insulin-like growth factor-1 (middle layer), and HA (deep layer). Results: The composite biological scaffold was conducive to adhesion, proliferation, and differentiation of mesenchymal stem cells recruited from the bone marrow and blood. Meanwhile, the environmental differences between the scaffold's layers contributed to the regional heterogeneity of chondrocytes and secreted proteins to promote functional cartilage regeneration. The biological effect of the composite scaffold was validated both in vitro and in vivo. Conclusion: A cartilage repair scaffold was established with high precision as well as promising mechanical and biological properties. This scaffold can promote the repair of cartilage injury by using, and inducing the differentiation and expression of, autologous bone marrow mesenchymal stem cells.
Collapse
|
28
|
Gao J, Zhang G, Xu K, Ma D, Ren L, Fan J, Hou J, Han J, Zhang L. Bone marrow mesenchymal stem cells improve bone erosion in collagen-induced arthritis by inhibiting osteoclasia-related factors and differentiating into chondrocytes. Stem Cell Res Ther 2020; 11:171. [PMID: 32381074 PMCID: PMC7203805 DOI: 10.1186/s13287-020-01684-w] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2019] [Revised: 04/06/2020] [Accepted: 04/20/2020] [Indexed: 12/12/2022] Open
Abstract
Background Rheumatoid arthritis (RA) is characterized by joint inflammation and damage to the cartilage and bone in collagen-induced arthritis (CIA). Mesenchymal stem cells (MSCs) can improve articular symptoms and reduce bone erosion in CIA rats; however, the underlying mechanism remains unknown. This study aimed to investigate the mechanism underlying MSC-induced improvement of bone destruction in CIA. Methods Wistar rats were divided into a normal group, CIA control group, MTX intervention group, and BMSC intervention group, each comprising 8 rats. Serum RANKL, OPG, and CXCL10 levels of all groups were determined via flow cytometry after 42 days of interventions. RANKL, OPG, TRAF6, CXCL10, and CXCR3 were detected on the synovial membrane via immunohistochemistry, and their relative mRNA levels were determined via RT-PCR analysis. BMSCs were labeled with GFP and administered to CIA rats via the tail vein. At different time points, the distribution of implanted GFP-MSCs in synovial tissues was observed using a fluorescence microscope, and the potential of GFP-MSCs to differentiate into chondrocytes was assessed via immunofluorescence analysis. Results BMSC transplantation improved joint inflammation and inhibited bone destruction in CIA rats. BMSCs inhibited the expression of serum CXCL10 and CXCL10 and CXCR3 expression at the synovial membrane. Moreover, protein and mRNA expression analyses revealed that BMSCs potentially regulated RANKL/OPG expression levels in the serum and synovial tissue. Upon implantation into CIA rats, GFP-MSCs were traced in the joints. GFP-positive cells were observed in the cartilage tissue from day 11 and until 42 days after transplantation. Anti-type II collagen/GFP double-positive cells were observed in the articular cartilage (especially damaged cartilage) upon immunofluorescence staining of anti-type II collagen. Conclusions BMSCs improve bone destruction in CIA by inhibiting the CXCL10/CXCR3 chemotactic axis, regulating the RANKL/OPG ratio, and directly differentiating into chondrocytes.
Collapse
Affiliation(s)
- Jinfang Gao
- Department of Rheumatology, Shanxi Bethune Hospital, Taiyuan, 030032, Shanxi, China.,Department of Rheumatology, Bethune Hospital Affiliated to Shanxi Medical University, Taiyuan, 030032, Shanxi, China
| | - Gailian Zhang
- Department of Rheumatology, Shanxi Bethune Hospital, Taiyuan, 030032, Shanxi, China
| | - Ke Xu
- Department of Rheumatology, Shanxi Bethune Hospital, Taiyuan, 030032, Shanxi, China
| | - Dan Ma
- Department of Rheumatology, Shanxi Bethune Hospital, Taiyuan, 030032, Shanxi, China.,Department of Rheumatology, Bethune Hospital Affiliated to Shanxi Medical University, Taiyuan, 030032, Shanxi, China
| | - Limin Ren
- Department of Rheumatology, Shanxi Bethune Hospital, Taiyuan, 030032, Shanxi, China
| | - Jingjing Fan
- Department of Rheumatology, Shanxi Bethune Hospital, Taiyuan, 030032, Shanxi, China
| | - Jianwen Hou
- Department of Rheumatology, Shanxi Bethune Hospital, Taiyuan, 030032, Shanxi, China
| | - Jian Han
- Department of Rheumatology, Shanxi Bethune Hospital, Taiyuan, 030032, Shanxi, China
| | - Liyun Zhang
- Department of Rheumatology, Shanxi Bethune Hospital, Taiyuan, 030032, Shanxi, China.
| |
Collapse
|
29
|
Sharifi F, Irani S, Azadegan G, Pezeshki-Modaress M, Zandi M, Saeed M. Co-electrospun gelatin-chondroitin sulfate/polycaprolactone nanofibrous scaffolds for cartilage tissue engineering. ACTA ACUST UNITED AC 2020. [DOI: 10.1016/j.bcdf.2020.100215] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
30
|
Irani S, Honarpardaz A, Choubini N, Pezeshki‐Modaress M, Zandi M. Chondro‐inductive nanofibrous scaffold based gelatin/polyvinyl alcohol/chondroitin sulfate for cartilage tissue engineering. POLYM ADVAN TECHNOL 2020. [DOI: 10.1002/pat.4869] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Shiva Irani
- Department of Biology, Science and Research BranchIslamic Azad University Tehran Iran
| | - Ali Honarpardaz
- Department of Biology, Science and Research BranchIslamic Azad University Tehran Iran
| | - Niloufar Choubini
- Department of Biology, Science and Research BranchIslamic Azad University Tehran Iran
| | | | - Mojgan Zandi
- Department of BiomaterialsIran Polymer and Petrochemical Institute Tehran Iran
| |
Collapse
|
31
|
Yilmaz EN, Zeugolis DI. Electrospun Polymers in Cartilage Engineering-State of Play. Front Bioeng Biotechnol 2020; 8:77. [PMID: 32133352 PMCID: PMC7039817 DOI: 10.3389/fbioe.2020.00077] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Accepted: 01/29/2020] [Indexed: 12/17/2022] Open
Abstract
Articular cartilage defects remain a clinical challenge. Articular cartilage defects progress to osteoarthritis, which negatively (e.g., remarkable pain, decreased mobility, distress) affects millions of people worldwide and is associated with excessive healthcare costs. Surgical procedures and cell-based therapies have failed to deliver a functional therapy. To this end, tissue engineering therapies provide a promise to deliver a functional cartilage substitute. Among the various scaffold fabrication technologies available, electrospinning is continuously gaining pace, as it can produce nano- to micro- fibrous scaffolds that imitate architectural features of native extracellular matrix supramolecular assemblies and can deliver variable cell populations and bioactive molecules. Herein, we comprehensively review advancements and shortfalls of various electrospun scaffolds in cartilage engineering.
Collapse
Affiliation(s)
- Elif Nur Yilmaz
- Regenerative, Modular & Developmental Engineering Laboratory, National University of Ireland Galway, Galway, Ireland.,Science Foundation Ireland, Centre for Research in Medical Devices, National University of Ireland Galway, Galway, Ireland
| | - Dimitrios I Zeugolis
- Regenerative, Modular & Developmental Engineering Laboratory, National University of Ireland Galway, Galway, Ireland.,Science Foundation Ireland, Centre for Research in Medical Devices, National University of Ireland Galway, Galway, Ireland
| |
Collapse
|
32
|
Olate-Moya F, Arens L, Wilhelm M, Mateos-Timoneda MA, Engel E, Palza H. Chondroinductive Alginate-Based Hydrogels Having Graphene Oxide for 3D Printed Scaffold Fabrication. ACS APPLIED MATERIALS & INTERFACES 2020; 12:4343-4357. [PMID: 31909967 DOI: 10.1021/acsami.9b22062] [Citation(s) in RCA: 83] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Scaffolds based on bioconjugated hydrogels are attractive for tissue engineering because they can partly mimic human tissue characteristics. For example, they can further increase their bioactivity with cells. However, most of the hydrogels present problems related to their processability, consequently limiting their use in 3D printing to produce tailor-made scaffolds. The goal of this work is to develop bioconjugated hydrogel nanocomposite inks for 3D printed scaffold fabrication through a micro-extrusion process having improved both biocompatibility and processability. The hydrogel is based on a photocrosslinkable alginate bioconjugated with both gelatin and chondroitin sulfate in order to mimic the cartilage extracellular matrix, while the nanofiller is based on graphene oxide to enhance the printability and cell proliferation. Our results show that the incorporation of graphene oxide into the hydrogel inks considerably improved the shape fidelity and resolution of 3D printed scaffolds because of a faster viscosity recovery post extrusion of the ink. Moreover, the nanocomposite inks produce anisotropic threads after the 3D printing process because of the templating of the graphene oxide liquid crystal. The in vitro proliferation assay of human adipose tissue-derived mesenchymal stem cells (hADMSCs) shows that bioconjugated scaffolds present higher cell proliferation than pure alginate, with the nanocomposites presenting the highest values at long times. Live/Dead assay otherwise displays full viability of hADMSCs adhered on the different scaffolds at day 7. Notably, the scaffolds produced with nanocomposite hydrogel inks were able to guide the cell proliferation following the direction of the 3D printed threads. In addition, the bioconjugated alginate hydrogel matrix induced chondrogenic differentiation without exogenous pro-chondrogenesis factors as concluded from immunostaining after 28 days of culture. This high cytocompatibility and chondroinductive effect toward hADMSCs, together with the improved printability and anisotropic structures, makes these nanocomposite hydrogel inks a promising candidate for cartilage tissue engineering based on 3D printing.
Collapse
Affiliation(s)
- Felipe Olate-Moya
- Departamento de Ingeniería Química, Biotecnología y Materiales, Facultad de Ciencias Físicas y Matemáticas , Universidad de Chile , Beauchef 851 , 8370456 Santiago , Chile
| | - Lukas Arens
- Institute for Technical Chemistry and Polymer Chemistry (ITCP) , Karlsruhe Institute of Technology (KIT) , Engesserstr. 18 , 76131 Karlsruhe , Germany
| | - Manfred Wilhelm
- Institute for Technical Chemistry and Polymer Chemistry (ITCP) , Karlsruhe Institute of Technology (KIT) , Engesserstr. 18 , 76131 Karlsruhe , Germany
| | - Miguel Angel Mateos-Timoneda
- Institute for Bioengineering of Catalonia (IBEC) , The Barcelona Institute of Science and Technology , Baldiri Reixac 10-12 , 08028 Barcelona , Spain
- CIBER en Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN) , Monforte de Lemos, 3-5 , 28029 Madrid , Spain
- Department of Materials Science, EEBE , Technical University of Catalonia (UPC) , d'Eduard Maristany 16 , 08019 Barcelona , Spain
| | - Elisabeth Engel
- Institute for Bioengineering of Catalonia (IBEC) , The Barcelona Institute of Science and Technology , Baldiri Reixac 10-12 , 08028 Barcelona , Spain
- CIBER en Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN) , Monforte de Lemos, 3-5 , 28029 Madrid , Spain
- Department of Materials Science, EEBE , Technical University of Catalonia (UPC) , d'Eduard Maristany 16 , 08019 Barcelona , Spain
| | - Humberto Palza
- Departamento de Ingeniería Química, Biotecnología y Materiales, Facultad de Ciencias Físicas y Matemáticas , Universidad de Chile , Beauchef 851 , 8370456 Santiago , Chile
- Millennium Nuclei in Soft Smart Mechanical Metamaterials , Beauchef 851 , 8370456 Santiago , Chile
| |
Collapse
|
33
|
W. King M, Chen J, Deshpande M, He T, Ramakrishna H, Xie Y, Zhang F, Zhao F. Structural Design, Fabrication and Evaluation of Resorbable Fiber-Based Tissue Engineering Scaffolds. Biotechnol Bioeng 2019. [DOI: 10.5772/intechopen.84643] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
34
|
Han Y, Li X, Zhang Y, Han Y, Chang F, Ding J. Mesenchymal Stem Cells for Regenerative Medicine. Cells 2019; 8:E886. [PMID: 31412678 PMCID: PMC6721852 DOI: 10.3390/cells8080886] [Citation(s) in RCA: 634] [Impact Index Per Article: 126.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 08/05/2019] [Accepted: 08/06/2019] [Indexed: 02/06/2023] Open
Abstract
In recent decades, the biomedical applications of mesenchymal stem cells (MSCs) have attracted increasing attention. MSCs are easily extracted from the bone marrow, fat, and synovium, and differentiate into various cell lineages according to the requirements of specific biomedical applications. As MSCs do not express significant histocompatibility complexes and immune stimulating molecules, they are not detected by immune surveillance and do not lead to graft rejection after transplantation. These properties make them competent biomedical candidates, especially in tissue engineering. We present a brief overview of MSC extraction methods and subsequent potential for differentiation, and a comprehensive overview of their preclinical and clinical applications in regenerative medicine, and discuss future challenges.
Collapse
Affiliation(s)
- Yu Han
- Department of Orthopedics, The Second Hospital of Jilin University, 218 Ziqiang Street, Changchun 130041, China
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun 130022, China
| | - Xuezhou Li
- Department of Orthopedics, The Second Hospital of Jilin University, 218 Ziqiang Street, Changchun 130041, China
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun 130022, China
| | - Yanbo Zhang
- Department of Orthopedics, China-Japan Union Hospital of Jilin University, 126 Xiantai Street, Changchun 130033, China.
| | - Yuping Han
- Department of Urology, China-Japan Union Hospital of Jilin University, 126 Xiantai Street, Changchun 130033, China.
| | - Fei Chang
- Department of Orthopedics, The Second Hospital of Jilin University, 218 Ziqiang Street, Changchun 130041, China.
| | - Jianxun Ding
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun 130022, China
| |
Collapse
|
35
|
Multi-Functional Electrospun Nanofibers from Polymer Blends for Scaffold Tissue Engineering. FIBERS 2019. [DOI: 10.3390/fib7070066] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Electrospinning and polymer blending have been the focus of research and the industry for their versatility, scalability, and potential applications across many different fields. In tissue engineering, nanofiber scaffolds composed of natural fibers, synthetic fibers, or a mixture of both have been reported. This review reports recent advances in polymer blended scaffolds for tissue engineering and the fabrication of functional scaffolds by electrospinning. A brief theory of electrospinning and the general setup as well as modifications used are presented. Polymer blends, including blends with natural polymers, synthetic polymers, mixture of natural and synthetic polymers, and nanofiller systems, are discussed in detail and reviewed.
Collapse
|
36
|
Tough, hybrid chondroitin sulfate nanofibers as a promising scaffold for skin tissue engineering. Int J Biol Macromol 2019; 132:63-75. [DOI: 10.1016/j.ijbiomac.2019.03.208] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2019] [Revised: 03/19/2019] [Accepted: 03/26/2019] [Indexed: 12/19/2022]
|
37
|
Chondroitin sulfate immobilized PCL nanofibers enhance chondrogenic differentiation of mesenchymal stem cells. Int J Biol Macromol 2019; 136:616-624. [PMID: 31207331 DOI: 10.1016/j.ijbiomac.2019.06.061] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 05/30/2019] [Accepted: 06/10/2019] [Indexed: 12/17/2022]
Abstract
Cold Atmospheric Plasma (CAP) is used as a promising method in surface modification for immobilization of chondroitin sulfate functional biomacromolecules on PCL nanofibrous substrates for cartilage tissue engineering. The GAG-grafted scaffolds are able to successfully support the attachment and proliferation of mesenchymal stem cells (MSCs). The seeded scaffolds show the chondro-differentiation of MSCs during a 21-days cell culture in a non-differential medium. Expression of SOX9, Collagen10 and Collagen2 proved the chondro-inductive effect of GAG-grafted scaffolds. Besides, no external chondro-genic differential agent was used in the differentiation of MSCs to chondrocyte. The cells passed the last phase of chondrogenesis after 14 days of incubation. Thus, the GAG-fabricated fibrous scaffolds using CAP are potential candidates for cartilage tissue engineering.
Collapse
|