1
|
Hayashi K, Tokumaru T, Shibahara K, Taleb Alashkar AN, Zhang C, Kishida R, Nakashima Y, Ishikawa K. Wood-Derived Hydrogels for Osteochondral Defect Repair. ACS NANO 2024. [PMID: 39730305 DOI: 10.1021/acsnano.4c10430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2024]
Abstract
Repairing cartilage tissue is a serious global challenge. Herein, we focus on wood skeletal structures that are highly porous for cell penetration yet have load-bearing strength, and aim to synthesize wood-derived hydrogels with the ability to regenerate cartilage tissues. The hydrogels were synthesized by wood delignification and the subsequent intercalation of citric acid (CA), which is involved in tricarboxylic acid cycles and essential for energy production, and N-acetylglucosamine (NAG), which is a cartilage glycosaminoglycan, among cellulose microfibrils. CA and NAG intercalation increased the amorphous region of the cellulose microfibrils and endowed them with flexibility while maintaining the skeletal structure of the wood. Consequently, the CA-NAG-treated wood hydrogels became twistable and bendable, and the acquired stiffness, compressive strength, water content, and cushioning characteristics were similar to those of the cartilage. In rabbit femur cartilage defects, CA-NAG-treated wood hydrogels induced the differentiation of surrounding cells into chondrocytes. Consequently, the CA-NAG-treated wood hydrogels repaired cartilage defects, whereas the collagen scaffolds, delignified wood materials, and CA-treated wood hydrogels did not. The CA-NAG-treated wood hydrogels exhibit superior structural and mechanical characteristics over conventional cellulose-fiber-containing scaffolds. Furthermore, the CA-NAG-treated wood hydrogels can effectively repair cartilage on their own, whereas conventional natural and synthetic polymeric materials need to be combined with cells and growth factors to achieve a sufficient therapeutic effect. Therefore, the CA-NAG-treated wood hydrogels successfully address the limitations of current therapies that either fail to repair articular cartilage or sacrifice healthy cartilage. To our knowledge, this is the pioneer study on the utilization of thinned wood for tissue engineering, which will contribute to solving both global health and environmental problems and to creating a sustainable society.
Collapse
Affiliation(s)
- Koichiro Hayashi
- Department of Biomaterials, Faculty of Dental Science, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Tatsuya Tokumaru
- Department of Biomaterials, Faculty of Dental Science, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Keigo Shibahara
- Department of Biomaterials, Faculty of Dental Science, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Ahmad Nazir Taleb Alashkar
- Department of Biomaterials, Faculty of Dental Science, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Cheng Zhang
- Department of Biomaterials, Faculty of Dental Science, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Ryo Kishida
- Department of Biomaterials, Faculty of Dental Science, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Yasuharu Nakashima
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Kunio Ishikawa
- Department of Biomaterials, Faculty of Dental Science, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| |
Collapse
|
2
|
Torkashvand M, Rezakhani L, Habibi Z, Mikaeili A, Rahmati S. Innovative approaches in lung tissue engineering: the role of exosome-loaded bioscaffolds in regenerative medicine. Front Bioeng Biotechnol 2024; 12:1502155. [PMID: 39758953 PMCID: PMC11695380 DOI: 10.3389/fbioe.2024.1502155] [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: 09/26/2024] [Accepted: 12/04/2024] [Indexed: 01/07/2025] Open
Abstract
Lung diseases account for over four million premature deaths every year, and experts predict that this number will increase in the future. The top cause of death globally is diseases which include conditions like lung cancer asthma and COPD. Treating severe acute lung injury is a complex task because lungs struggle to heal themselves in the presence of swelling inflammation and scarring caused by damage, to the lung tissues. Though achieving lung regeneration, in controlled environments is still an ambition; ongoing studies are concentrating on notable progress, in the field of lung tissue engineering and methods for repairing lung damage. This review delves into methods, for regenerating lungs with a focus on exosome carry bioscaffolds and mesenchymal stem cells among others. It talks about how these new techniques can help repair lung tissue and improve lung function in cases of damage. Also noted is the significance of ex vivo lung perfusion (EVLP), for rejuvenating donor lungs and the healing properties of exosomes in supporting lung regeneration.
Collapse
Affiliation(s)
| | - Leila Rezakhani
- Fertility and Infertility Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
- Department of Tissue Engineering, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Zahra Habibi
- Clinical Research Development Unit, Hajar Hospital, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Abdolhamid Mikaeili
- Medical Biology Research Center, Health Technology Institute, University of Medical Sciences, Kermanshah, Iran
| | - Shima Rahmati
- Cancer Research Center, Shahrekord University of Medical Sciences, Shahrekord, Iran
| |
Collapse
|
3
|
Mazetyte-Godiene A, Vailionyte A, Jelinskas T, Denkovskij J, Usas A. Promotion of hMDSC differentiation by combined action of scaffold material and TGF-β superfamily growth factors. Regen Ther 2024; 27:307-318. [PMID: 38633416 PMCID: PMC11021853 DOI: 10.1016/j.reth.2024.03.018] [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: 08/03/2023] [Revised: 01/08/2024] [Accepted: 03/17/2024] [Indexed: 04/19/2024] Open
Abstract
Objective Herein we propose a combined action of collagen type I (CA) or synthetic collagen-like-peptide functionalized with the cell adhesive RGD motif (PEG-CLP-RGD) hydrogels and selected growth factors to promote chondrogenic differentiation of human muscle-derived stem cells (hMDSCs) under normal and reduced oxygen conditions. Methods hMDSCs were set for differentiation towards chondrogenic lineage using BMP-7 and TGF-β3. Cells were seeded onto hydrogels loaded with growth factors (75ng/scaffold) and cultured for 28 days under normal (21%) and severe hypoxic (1%) conditions. Chondrogenesis was evaluated by monitoring collagen type II and GAG deposition, and quantification of ACAN expression by RT-PCR. Results Sustained release of TGFβ3 from the hydrogels was observed, 8.7 ± 0.5% of the initially loaded amount diffused out after 24 h from both substrates. For the BMP-7 growth factor, 14.8 ± 0.3% and 18.2 ± 0.6% of the initially loaded amount diffused out after 24 h from CA and CLP-RGD, respectively. The key findings of this study are: i) the self-supporting hydrogels themselves can stimulate hMDSC chondrogenesis by inducing gene expression of cartilage-specific proteoglycan aggrecan and ECM production; ii) the effect of dual BMP-7 and TGF-β3 loading was more pronounced on CA hydrogel under normal oxygen conditions; iii) dual loading on PEG-CLP-RGD hydrogels did not have the synergistic effect, TGF-β3 was more effective under both oxygen conditions; iv) BMP-7 can improve chondrogenic effect of TGF-β3 on CA scaffolds, and hydrogels loaded with both growth factors can induce cartilage formation in hMDSC cultures. Conclusion Our results support the potential strategy of combining implantable hydrogels functionalized with differentiation factors toward improving cartilaginous repair.
Collapse
Affiliation(s)
- Airina Mazetyte-Godiene
- Institute of Physiology and Pharmacology, Medical Academy, Lithuanian University of Health Sciences, Kaunas, Lithuania
- UAB Ferentis, Savanoriu ave. 235, Vilnius, Lithuania
- Department of Nanoengineering, Center for Physical Sciences and Technology, Vilnius, Lithuania
| | | | - Tadas Jelinskas
- Department of Nanoengineering, Center for Physical Sciences and Technology, Vilnius, Lithuania
| | - Jaroslav Denkovskij
- Department of Regenerative Medicine, Centre for Innovative Medicine, Vilnius, Lithuania
| | - Arvydas Usas
- Institute of Physiology and Pharmacology, Medical Academy, Lithuanian University of Health Sciences, Kaunas, Lithuania
| |
Collapse
|
4
|
Rafiq M, Ahmed J, Alturaifi HA, Awwad NS, Ibrahium HA, Mir S, Maalik A, Sabahat S, Hassan S, Khan ZUH. Recent developments in the biomedical and anticancer applications of chitosan derivatives. Int J Biol Macromol 2024; 283:137601. [PMID: 39549805 DOI: 10.1016/j.ijbiomac.2024.137601] [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: 08/09/2024] [Revised: 11/04/2024] [Accepted: 11/11/2024] [Indexed: 11/18/2024]
Abstract
Chitosan is a natural polymer derived from chitin. It has significant applications in various fields due to its unique physicochemical properties, biocompatibility, and biodegradability. These important properties of chitosan make it an attractive candidate for various anti-cancer activities and biomedical applications, including tissue engineering. This review emphasizes the latest literature on anticancer applications of chitosan derivatives and in-depth study of biomedical applications. This review highlights the importance of biomedical applications and anti-cancer activities like breast, liver, colon, gastric, melanoma, colorectal, cervical, oral, and lymphoma cancer. Currently, there is a notable absence of recent reviews that comprehensively address these aspects such as Alejandro Elizalde-Cárdenas, et al. 2024, focuses only on Biomedical applications of Cs and its derivatives (Elizalde-Cárdenas et al., 2024). Jingxian Ding, et al. 2022 discussed the applications of Cs in some Cancer treatments (Mabrouk et al., 2024). However, our article aims to provide a comprehensive overview of the latest advancements in Cs derivatives in both fields. This manuscript is designed with proper diagrams, flow sheets and summarized tables to enhance the understanding of the reader. It also highlights recent advancements in the development of various chitosan derivatives, offering a comprehensive perspective for researchers and practitioners to further progress in biomedical and anticancer technologies.
Collapse
Affiliation(s)
- Muqadas Rafiq
- Department of Chemistry, COMSATS University Islamabad, Islamabad Campus, Park Road, Chak Shahzad, Islamabad 44000, Pakistan
| | - Jalal Ahmed
- Department of Chemistry, COMSATS University Islamabad, Islamabad Campus, Park Road, Chak Shahzad, Islamabad 44000, Pakistan
| | - Huriyyah A Alturaifi
- Chemistry Department, Faculty of Science, King Khalid University, PO Box 9004, Abha 61413, Saudi Arabia
| | - Nasser S Awwad
- Chemistry Department, Faculty of Science, King Khalid University, PO Box 9004, Abha 61413, Saudi Arabia
| | - Hala A Ibrahium
- Biology Department, Faculty of Science, King Khalid University, PO Box 9004, Abha 61413, Saudi Arabia
| | - Sadullah Mir
- Department of Chemistry, COMSATS University Islamabad, Islamabad Campus, Park Road, Chak Shahzad, Islamabad 44000, Pakistan.
| | - Aneela Maalik
- Department of Chemistry, COMSATS University Islamabad, Islamabad Campus, Park Road, Chak Shahzad, Islamabad 44000, Pakistan
| | - Sana Sabahat
- Department of Chemistry, COMSATS University Islamabad, Islamabad Campus, Park Road, Chak Shahzad, Islamabad 44000, Pakistan
| | - Safia Hassan
- Department of Chemistry, COMSATS University Islamabad, Islamabad Campus, Park Road, Chak Shahzad, Islamabad 44000, Pakistan
| | - Zia Ul Haq Khan
- Department of Chemistry, COMSATS University Islamabad, Islamabad Campus, Park Road, Chak Shahzad, Islamabad 44000, Pakistan
| |
Collapse
|
5
|
Plath AMS, de Lima PHC, Amicone A, Bissacco EG, Mosayebi M, Berton SBR, Ferguson SJ. Toward low-friction and high-adhesion solutions: Emerging strategies for nanofibrous scaffolds in articular cartilage engineering. BIOMATERIALS ADVANCES 2024; 169:214129. [PMID: 39642717 DOI: 10.1016/j.bioadv.2024.214129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Revised: 11/28/2024] [Accepted: 11/29/2024] [Indexed: 12/09/2024]
Abstract
Aging, trauma, pathology, and poor natural tissue regeneration are the leading causes of osteoarthritis (OA), an articular cartilage disease. Electrospun scaffolds have gained attention as potential matrices for the treatment of OA because of their high degree of ECM mimicry, which suits chondrocyte migration, adhesion, and proliferation. However, none of the products recently introduced in the market are nanofiber-based. This study aimed to review the scope and tribology of nanofibrous articular cartilage scaffolds. Herein, we briefly discuss cartilage lubrication and strategies for promoting cell adhesion in electrospun materials. Next, we discuss the emerging need to study the biotribological properties of scaffolds. Finally, we review new perspectives on surface functionalization, surface segregation, Janus membranes, layer-by-layer fabrication, and nanofibrous composites. We conclude that cell adhesion and low-friction conciliation remain poorly explored in the recent literature. The topic intersection might create novelties in the field.
Collapse
Affiliation(s)
| | - Pedro Henrique Correia de Lima
- Department of Physics and Chemistry, São Paulo State University (UNESP), Av. Brasil, 56, 15385007 Ilha Solteira, Brazil.
| | - Alessio Amicone
- Institute for Biomechanics, ETH Zurich, Gloriastrasse 37-39, 8092 Zurich, Switzerland
| | | | - Mahdieh Mosayebi
- Institute for Biomechanics, ETH Zurich, Gloriastrasse 37-39, 8092 Zurich, Switzerland
| | | | - Stephen J Ferguson
- Institute for Biomechanics, ETH Zurich, Gloriastrasse 37-39, 8092 Zurich, Switzerland
| |
Collapse
|
6
|
Effanga VE, Akilbekova D, Mukasheva F, Zhao X, Kalyon DM, Erisken C. In Vitro Investigation of 3D Printed Hydrogel Scaffolds with Electrospun Tidemark Component for Modeling Osteochondral Interface. Gels 2024; 10:745. [PMID: 39590101 PMCID: PMC11593412 DOI: 10.3390/gels10110745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Revised: 10/23/2024] [Accepted: 11/13/2024] [Indexed: 11/28/2024] Open
Abstract
Osteochondral (OC) tissue plays a crucial role due to its ability to connect bone and cartilage tissues. To address the complexity of structure and functionality at the bone-cartilage interface, relevant to the presence of the tidemark as a critical element at the bone-cartilage boundary, we fabricated graded scaffolds through sequential 3D printing. The scaffold's bottom layer was based on a gelatin/oxidized alginate mixture enriched with hydroxyapatite (HAp) to create a rougher surface and larger pores to promote osteogenesis. In contrast, the upper layer was engineered to have smaller pores and aimed to promote cartilage tissue formation and mimic the physical properties of the cartilage. An electrospun ε-polycaprolactone (PCL) membrane with micrometer-range pores was incorporated between the layers to replicate the function of tidemark-a barrier to prevent vascularization of cartilage from subchondral bone tissue. In vitro cell studies confirmed the viability of the cells on the layers of the scaffolds and the ability of PCL mesh to prevent cellular migration. The fabricated scaffolds were thoroughly characterized, and their mechanical properties were compared to native OC tissue, demonstrating suitability for OC tissue engineering and graft modeling. The distance of gradient of mineral concentration was found to be 151 µm for grafts and the native OC interface.
Collapse
Affiliation(s)
- Victoria Effiong Effanga
- Department of Chemical and Materials Engineering, School of Engineering and Digital Sciences, Nazarbayev University, 010000 Astana, Kazakhstan; (V.E.E.); (D.A.); (F.M.)
| | - Dana Akilbekova
- Department of Chemical and Materials Engineering, School of Engineering and Digital Sciences, Nazarbayev University, 010000 Astana, Kazakhstan; (V.E.E.); (D.A.); (F.M.)
| | - Fariza Mukasheva
- Department of Chemical and Materials Engineering, School of Engineering and Digital Sciences, Nazarbayev University, 010000 Astana, Kazakhstan; (V.E.E.); (D.A.); (F.M.)
| | - Xiao Zhao
- Department of Chemical Engineering and Material Science, Stevens Institute of Technology, Hoboken, NJ 07030, USA; (X.Z.); (D.M.K.)
| | - Dilhan M. Kalyon
- Department of Chemical Engineering and Material Science, Stevens Institute of Technology, Hoboken, NJ 07030, USA; (X.Z.); (D.M.K.)
| | - Cevat Erisken
- Department of Chemical and Materials Engineering, School of Engineering and Digital Sciences, Nazarbayev University, 010000 Astana, Kazakhstan; (V.E.E.); (D.A.); (F.M.)
| |
Collapse
|
7
|
Asadikorayem M, Brunel LG, Weber P, Heilshorn SC, Zenobi-Wong M. Porosity dominates over microgel stiffness for promoting chondrogenesis in zwitterionic granular hydrogels. Biomater Sci 2024; 12:5504-5520. [PMID: 39347711 PMCID: PMC11441418 DOI: 10.1039/d4bm00233d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Accepted: 09/15/2024] [Indexed: 10/01/2024]
Abstract
Granular hydrogels comprised of jammed, crosslinked microgels offer great potential as biomaterial scaffolds for cell-based therapies, including for cartilage tissue regeneration. As stiffness and porosity of hydrogels affect the phenotype of encapsulated cells and the extent of tissue regeneration, the design of tunable granular hydrogels to control and optimize these parameters is highly desirable. We hypothesized that chondrogenesis could be modulated using a granular hydrogel platform based on biocompatible, zwitterionic materials with independent intra- and inter-microgel crosslinking mechanisms. Microgels are made with mechanical fragmentation of photocrosslinked zwitterionic carboxybetaine acrylamide (CBAA) and sulfobetaine methacrylate (SBMA) hydrogels, and secondarily crosslinked in the presence of cells using horseradish peroxide (HRP) to produce cell-laden granular hydrogels. We varied the intra-microgel crosslinking density to produce microgels with varied stiffnesses (1-3 kPa) and swelling properties. These microgels, when resuspended at the same weight fraction and secondarily crosslinked, resulted in granular hydrogels with distinct porosities (5-40%) due to differing swelling properties. The greatest extent of chondrogenesis was achieved in scaffolds with the highest microgel stiffness and highest porosity. However, when scaffold porosity was kept constant and just microgel stiffness varied, cell phenotype and chondrogenesis were similar across scaffolds. These results indicate the dominant role of granular scaffold porosity on chondrogenesis, whereas microgel stiffness appears to play a relatively minor role. These observations are in contrast to cells encapsulated within conventional bulk hydrogels, where stiffness has been shown to significantly affect chondrocyte response. In summary, we introduce chemically-defined, zwitterionic biomaterials to fabricate versatile granular hydrogels allowing for tunable scaffold porosity and microgel stiffness to study and influence chondrogenesis.
Collapse
Affiliation(s)
- Maryam Asadikorayem
- Department of Health Sciences and Technology, ETH Zürich, Zürich, Switzerland.
| | - Lucia G Brunel
- Department of Chemical Engineering, Stanford University, Stanford, CA, USA
| | - Patrick Weber
- Department of Health Sciences and Technology, ETH Zürich, Zürich, Switzerland.
| | - Sarah C Heilshorn
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, USA
| | - Marcy Zenobi-Wong
- Department of Health Sciences and Technology, ETH Zürich, Zürich, Switzerland.
| |
Collapse
|
8
|
Omrani E, Haramshahi MA, Najmoddin N, Saeed M, Pezeshki-Modaress M. Acceleration of chondrogenic differentiation utilizing biphasic core-shell alginate sulfate electrospun nanofibrous scaffold. Colloids Surf B Biointerfaces 2024; 242:114080. [PMID: 39003847 DOI: 10.1016/j.colsurfb.2024.114080] [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: 05/09/2024] [Revised: 07/04/2024] [Accepted: 07/06/2024] [Indexed: 07/16/2024]
Abstract
Engineering new biomedical materials with tailored physicochemical, mechanical and biological virtues in order to differentiate stem cells into chondrocytes for cartilage regeneration has garnered much scientific interest. In this study, core/shell nanofibrous scaffold based on poly(ɛ-caprolactone) (PCL) as a core material and alginate sulfate (AlgS)-poly(vinyl alcohol) (PVA) blend as shell materials (AlgS-PVA/PCL) was fabricated by emulsion electrospinning. In this vein, the influence of AlgS to PVA ratio (30:70, 50:50), organic to aqueous phase ratio (1:2, 1:3 and 1:5) and acid concentration (0, 10, 20, 30, 40 and 50 %) on nanofibers morphology were investigated. SEM images depicted that AlgS to PVA ratio of 30:70 and 50:50, organic to aqueous phase ratio of 1:3 and 1:5 and acid concentration of 30 % led to uniform, bead-free fibrous mats. AlgS-PVA/PCL scaffolds with AlgS to PVA ratio of 30:70 and organic to aqueous phase ratio of 1:3, showed admirable mechanical features, high porosity (>90 %) with desirable swelling ratio in wet condition. In vitro assays indicated that the AlgS-PVA/PCL scaffold surface had desirable interaction with stem cells and promotes cells attachment, proliferation and differentiation. Thus, we envision that this salient structure could be an intriguing construction as a cartilage tissue-engineered scaffold.
Collapse
Affiliation(s)
- Elmira Omrani
- Department of Biomedical Engineering, Medical Engineering and Biology Research Center, Science and Research Branch, Islamic Azad University, Tehran, the Islamic Republic of Iran
| | - Mohammad Amin Haramshahi
- Department of Tissue Engineering and Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, the Islamic Republic of Iran
| | - Najmeh Najmoddin
- Department of Biomedical Engineering, Medical Engineering and Biology Research Center, Science and Research Branch, Islamic Azad University, Tehran, the Islamic Republic of Iran.
| | - Mahdi Saeed
- Soft Tissue Engineering Research Center, Tissue Engineering and Regenerative Medicine Institute, Central Tehran Branch, Islamic Azad University, Tehran, the Islamic Republic of Iran
| | - Mohamad Pezeshki-Modaress
- Burn Research Center, Iran University of Medical Sciences, Tehran, the Islamic Republic of Iran; Department of Plastic and Reconstructive Surgery, Hazrat Fatemeh Hospital, School of Medicine, Iran University of Medical Sciences, Tehran, the Islamic Republic of Iran; Stem Cell and Regenerative Medicine Research Center, Iran University of Medical Sciences, Tehran, the Islamic Republic of Iran.
| |
Collapse
|
9
|
Jeyaraman N, Jeyaraman M, Muthu S, Balaji S, Ramasubramanian S, Patro BP. Chondrogenic Potential of Umbilical Cord-Derived Mesenchymal Stromal Cells: Insights and Innovations. Indian J Orthop 2024; 58:1349-1361. [PMID: 39324097 PMCID: PMC11420429 DOI: 10.1007/s43465-024-01239-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Accepted: 08/14/2024] [Indexed: 09/11/2024]
Abstract
Background The advent of tissue engineering and regenerative medicine has introduced innovative approaches to treating degenerative and traumatic injuries, particularly in cartilage, a tissue with limited self-repair capabilities. Among the various stem cell sources, umbilical cord-derived mesenchymal stromal cells (UC-MSCs) have garnered significant interest due to their non-invasive collection, minimal ethical concerns, and robust regenerative potential, particularly in cartilage regeneration. Methods A comprehensive literature review was conducted using multiple databases, including PubMed, Scopus, Web of Science, and Google Scholar. Search terms focused on "umbilical cordderived mesenchymal stromal cells," "chondrogenesis," "cartilage regeneration," and related topics. Studies published in the past two decades were included, with selection criteria emphasizing methodological rigor and relevance to UC-MSC chondrogenesis. The review synthesizes findings from various sources to provide a thorough analysis of the potential of UC-MSCs in cartilage tissue engineering. Results UC-MSCs exhibit significant chondrogenic potential, supported by their ability to differentiate into chondrocytes under specific conditions. Recent advancements include the development of biomaterial scaffolds and the application of genetic engineering techniques, such as CRISPR/Cas9, to enhance chondrogenic differentiation. Despite these advancements, challenges remain in standardizing cell isolation techniques, scaling up production for clinical use, and ensuring the long-term functionality of regenerated cartilage. Conclusion UC-MSCs offer a promising solution for cartilage regeneration in the field of regenerative medicine. Ongoing research is focused on overcoming current challenges through the use of advanced technologies, including bioreactors and gene editing. Collaborative efforts among researchers, clinicians, and bioengineers are essential to translating the potential of UC-MSCs into effective clinical therapies, which could significantly advance tissue regeneration and therapeutic innovation. Graphical Abstract
Collapse
Affiliation(s)
- Naveen Jeyaraman
- Department of Orthopaedics, ACS Medical College and Hospital, Dr MGR Educational and Research Institute, Tamil Nadu, Chennai, 600077 India
| | - Madhan Jeyaraman
- Department of Orthopaedics, ACS Medical College and Hospital, Dr MGR Educational and Research Institute, Tamil Nadu, Chennai, 600077 India
- VirginiaTech India, Dr MGR Educational and Research Institute, Tamil Nadu, Chennai, 600095 India
- Department of Orthopaedics, Orthopaedic Research Group, Tamil Nadu, Coimbatore, 641045 India
| | - Sathish Muthu
- Department of Orthopaedics, Orthopaedic Research Group, Tamil Nadu, Coimbatore, 641045 India
- Department of Biotechnology, Faculty of Engineering, Karpagam Academy of Higher Education, Tamil Nadu, Coimbatore, 641021 India
- Department of Orthopaedics, Government Karur Medical College, Tamil Nadu, Karur, 639004 India
| | - Sangeetha Balaji
- Department of Orthopaedics, Government Medical College, Omandurar Government Estate, Tamil Nadu, Chennai, 600002 India
| | - Swaminathan Ramasubramanian
- Department of Orthopaedics, Government Medical College, Omandurar Government Estate, Tamil Nadu, Chennai, 600002 India
| | - Bishnu Prasad Patro
- Department of Orthopaedics, All India Institute of Medical Sciences, Bhubaneswar, Odisha 751019 India
| |
Collapse
|
10
|
Yuan L, Yao L, Ren X, Chen X, Li X, Xu Y, Jin T. Cartilage defect repair in a rat model via a nanocomposite hydrogel loaded with melatonin-loaded gelatin nanofibers and menstrual blood stem cells: an in vitro and in vivo study. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2024; 35:55. [PMID: 39347832 PMCID: PMC11442572 DOI: 10.1007/s10856-024-06820-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Accepted: 07/26/2024] [Indexed: 10/01/2024]
Abstract
Cartilage damage caused by injuries or degenerative diseases remains a major challenge in the field of regenerative medicine. In this study, we developed a composite hydrogel system for the delivery of melatonin and menstrual blood stem cells (MenSCs) to treat a rat model of cartilage defect. The composite delivery system was produced by incorporation of melatonin into the gelatin fibers and dispersing these fibers into calcium alginate hydrogels. Various characterization methods including cell viability assay, microstructure studies, degradation rate measurement, drug release, anti-inflammatory assay, and radical scavenging assay were used to characterize the hydrogel system. MenSCs were encapsulated within the nanocomposite hydrogel and implanted into a rat model of full-thickness cartilage defect. A 1.3 mm diameter drilled in the femoral trochlea and used for the in vivo study. Results showed that the healing potential of nanocomposite hydrogels containing melatonin and MenSCs was significantly higher than polymer-only hydrogels. Our study introduces a novel composite hydrogel system, combining melatonin and MenSCs, demonstrating enhanced cartilage repair efficacy, offering a promising avenue for regenerative medicine.
Collapse
Affiliation(s)
- Libo Yuan
- Department of Orthopedics, 920 Hospital of Joint Logistic Support Force, Kunming, China
| | - Ling Yao
- Department of Orthopedics, 920 Hospital of Joint Logistic Support Force, Kunming, China
| | - Xianzhen Ren
- Department of Orthopedics, 920 Hospital of Joint Logistic Support Force, Kunming, China
| | - Xusheng Chen
- Department of Orthopedics, 920 Hospital of Joint Logistic Support Force, Kunming, China
| | - Xu Li
- Department of Orthopedics, 920 Hospital of Joint Logistic Support Force, Kunming, China
| | - Yongqing Xu
- Department of Orthopedics, 920 Hospital of Joint Logistic Support Force, Kunming, China.
| | - Tao Jin
- Department of Orthopedics, 920 Hospital of Joint Logistic Support Force, Kunming, China.
| |
Collapse
|
11
|
Karami P, Laurent A, Philippe V, Applegate LA, Pioletti DP, Martin R. Cartilage Repair: Promise of Adhesive Orthopedic Hydrogels. Int J Mol Sci 2024; 25:9984. [PMID: 39337473 PMCID: PMC11432485 DOI: 10.3390/ijms25189984] [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: 08/08/2024] [Revised: 09/09/2024] [Accepted: 09/13/2024] [Indexed: 09/30/2024] Open
Abstract
Cartilage repair remains a major challenge in human orthopedic medicine, necessitating the application of innovative strategies to overcome existing technical and clinical limitations. Adhesive hydrogels have emerged as promising candidates for cartilage repair promotion and tissue engineering, offering key advantages such as enhanced tissue integration and therapeutic potential. This comprehensive review navigates the landscape of adhesive hydrogels in cartilage repair, discussing identified challenges, shortcomings of current treatment options, and unique advantages of adhesive hydrogel products and scaffolds. While emphasizing the critical need for in situ lateral integration with surrounding tissues, we dissect current limitations and outline future perspectives for hydrogel scaffolds in cartilage repair. Moreover, we examine the clinical translation pathway and regulatory considerations specific to adhesive hydrogels. Overall, this review synthesizes the existing insights and knowledge gaps and highlights directions for future research regarding adhesive hydrogel-based devices in advancing cartilage tissue engineering.
Collapse
Affiliation(s)
- Peyman Karami
- Department of Orthopedic Surgery and Traumatology, University Hospital of Lausanne, CH-1011 Lausanne, Switzerland
- Laboratory of Biomechanical Orthopaedics, Institute of Bioengineering, School of Engineering, EPFL, CH-1015 Lausanne, Switzerland
| | - Alexis Laurent
- Manufacturing Department, LAM Biotechnologies SA, CH-1066 Epalinges, Switzerland
- Regenerative Therapy Unit, Reconstructive and Hand Surgery Service, Lausanne University Hospital, University of Lausanne, CH-1066 Epalinges, Switzerland
| | - Virginie Philippe
- Department of Orthopedic Surgery and Traumatology, University Hospital of Lausanne, CH-1011 Lausanne, Switzerland
- Regenerative Therapy Unit, Reconstructive and Hand Surgery Service, Lausanne University Hospital, University of Lausanne, CH-1066 Epalinges, Switzerland
| | - Lee Ann Applegate
- Regenerative Therapy Unit, Reconstructive and Hand Surgery Service, Lausanne University Hospital, University of Lausanne, CH-1066 Epalinges, Switzerland
- Center for Applied Biotechnology and Molecular Medicine, University of Zurich, CH-8057 Zurich, Switzerland
- Oxford OSCAR Suzhou Center, Oxford University, Suzhou 215123, China
| | - Dominique P Pioletti
- Laboratory of Biomechanical Orthopaedics, Institute of Bioengineering, School of Engineering, EPFL, CH-1015 Lausanne, Switzerland
| | - Robin Martin
- Department of Orthopedic Surgery and Traumatology, University Hospital of Lausanne, CH-1011 Lausanne, Switzerland
| |
Collapse
|
12
|
Jakutowicz T, Wasyłeczko M, Płończak M, Wojciechowski C, Chwojnowski A, Czubak J. Comparative Study of Autogenic and Allogenic Chondrocyte Transplants on Polyethersulfone Scaffolds for Cartilage Regeneration. Int J Mol Sci 2024; 25:9075. [PMID: 39201763 PMCID: PMC11354243 DOI: 10.3390/ijms25169075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 08/10/2024] [Accepted: 08/19/2024] [Indexed: 09/03/2024] Open
Abstract
The aim of this study was to evaluate the chondrogenic potential of chondrocyte transplants cultured in vitro on polyethersulfone (PES) membranes. Forty-eight rabbits (96 knee joints) were used in the project. The synthetic, macro-porous PES membranes were used as scaffolds. Fragments of articular cartilage were harvested from non-weight-bearing areas of the joints of the animals. Chondrocytes were isolated and then cultivated on PES scaffolds for 3 weeks. The animals were divided into four groups. All the lesions in the articular cartilage were full thickness defects. In Group I, autogenic chondrocytes on PES membranes were transplanted into the defect area; in Group II, allogenic chondrocytes on PES membranes were transplanted into the defect area; in Group III, pure PES membranes were transplanted into the defect area; and in Group IV, lesions were left untreated. Half of the animals from each group were terminated after 8 weeks, and the remaining half were terminated 12 weeks postoperatively. The samples underwent macroscopic evaluation using the Brittberg scale and microscopic evaluation using the O'Driscoll scale. The best regeneration was observed in Groups II and I. In Group I, the results were achieved with two surgeries, while in Group II, only one operation was needed. This indicates that allogenic chondrocytes do not require two surgeries, highlighting the importance of further in vivo studies to better understand this advantage. The success of the study and the desired properties of PES scaffolds are attributed mainly to the presence of sulfonic groups in the structure of the material. These groups, similar to chondroitin sulfate, which naturally occurs in hyaline cartilage, likely enable mutual affinity between the scaffold and cells and promote scaffold colonization by the cells.
Collapse
Affiliation(s)
- Tomasz Jakutowicz
- Paediatric Orthopaedics and Traumatology Department, Children’s Hospital, Medical University of Warsaw, 02-091 Warsaw, Poland
| | - Monika Wasyłeczko
- Nałęcz Institute of Biocybernetics and Biomedical Engineering Polish Academy of Sciences, 02-109 Warsaw, Poland
| | - Maciej Płończak
- Department of Orthopedics, Medical Centre of Postgraduate Education, 01-813 Warsaw, Poland
- Gruca Teaching Hospital, 05-400 Otwock, Poland
- Mazovia Regional Hospital in Siedlce, 08-110 Siedlce, Poland
| | - Cezary Wojciechowski
- Nałęcz Institute of Biocybernetics and Biomedical Engineering Polish Academy of Sciences, 02-109 Warsaw, Poland
| | - Andrzej Chwojnowski
- Nałęcz Institute of Biocybernetics and Biomedical Engineering Polish Academy of Sciences, 02-109 Warsaw, Poland
| | - Jarosław Czubak
- Department of Orthopedics, Medical Centre of Postgraduate Education, 01-813 Warsaw, Poland
- Gruca Teaching Hospital, 05-400 Otwock, Poland
| |
Collapse
|
13
|
Gao S, Nie T, Lin Y, Jiang L, Wang L, Wu J, Jiao Y. 3D printing tissue-engineered scaffolds for auricular reconstruction. Mater Today Bio 2024; 27:101141. [PMID: 39045312 PMCID: PMC11265588 DOI: 10.1016/j.mtbio.2024.101141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 06/24/2024] [Accepted: 06/28/2024] [Indexed: 07/25/2024] Open
Abstract
Congenital microtia is the most common cause of auricular defects, with a prevalence of approximately 5.18 per 10,000 individuals. Autologous rib cartilage grafting is the leading treatment modality at this stage of auricular reconstruction currently. However, harvesting rib cartilage may lead to donor site injuries, such as pneumothorax, postoperative pain, chest wall scarring, and deformity. Therefore, in the pursuit of better graft materials, biomaterial scaffolds with great histocompatibility, precise control of morphology, non-invasiveness properties are gradually becoming a new research hotspot in auricular reconstruction. This review collectively presents the exploit and application of 3D printing biomaterial scaffold in auricular reconstruction. Although the tissue-engineered ear still faces challenges before it can be widely applied to patients in clinical settings, and its long-term effects have yet to be evaluated, we aim to provide guidance for future research directions in 3D printing biomaterial scaffold for auricular reconstruction. This will ultimately benefit the translational and clinical application of cartilage tissue engineering and biomaterials in the treatment of auricular defects.
Collapse
Affiliation(s)
- Shuyi Gao
- Department of Otorhinolaryngology Head and Neck Surgery, Guangzhou Twelfth People's Hospital (The Affiliated Twelfth People's Hospital of Guangzhou Medical University), Guangzhou Medical University, Guangzhou, 510620, China
- Institute of Otorhinolaryngology, Head and Neck Surgery, Guangzhou Medical University, Guangzhou, 510620, China
| | - Tianqi Nie
- Department of Otorhinolaryngology Head and Neck Surgery, Guangzhou Twelfth People's Hospital (The Affiliated Twelfth People's Hospital of Guangzhou Medical University), Guangzhou Medical University, Guangzhou, 510620, China
- Institute of Otorhinolaryngology, Head and Neck Surgery, Guangzhou Medical University, Guangzhou, 510620, China
| | - Ying Lin
- Department of Otolaryngology Head and Neck Surgery, Guangzhou Red Cross Hospital (Guangzhou Red Cross Hospital of Jinan University), Jinan University, Guangzhou, 510240, China
- Institute of Otolaryngology Head and Neck Surgery, Jinan University, Guangzhou, 510240, China
| | - Linlan Jiang
- Department of Otorhinolaryngology Head and Neck Surgery, Guangzhou Twelfth People's Hospital (The Affiliated Twelfth People's Hospital of Guangzhou Medical University), Guangzhou Medical University, Guangzhou, 510620, China
- Institute of Otorhinolaryngology, Head and Neck Surgery, Guangzhou Medical University, Guangzhou, 510620, China
| | - Liwen Wang
- Department of Otorhinolaryngology Head and Neck Surgery, Guangzhou Twelfth People's Hospital (The Affiliated Twelfth People's Hospital of Guangzhou Medical University), Guangzhou Medical University, Guangzhou, 510620, China
- Institute of Otorhinolaryngology, Head and Neck Surgery, Guangzhou Medical University, Guangzhou, 510620, China
| | - Jun Wu
- Institute of Otorhinolaryngology, Head and Neck Surgery, Guangzhou Medical University, Guangzhou, 510620, China
- Bioscience and Biomedical Engineering Thrust, The Hong Kong University of Science and Technology (Guangzhou), Nansha, Guangzhou, 511400, China
- Division of Life Science, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Yuenong Jiao
- Department of Otorhinolaryngology Head and Neck Surgery, Guangzhou Twelfth People's Hospital (The Affiliated Twelfth People's Hospital of Guangzhou Medical University), Guangzhou Medical University, Guangzhou, 510620, China
- Institute of Otorhinolaryngology, Head and Neck Surgery, Guangzhou Medical University, Guangzhou, 510620, China
| |
Collapse
|
14
|
Hasan SMK, Islam SR, Zerin I, Ahmed T, Rahman S. Gelatin/EGDE Ultrafine Composite Fibers Reinforced with 3D Spacer Fabric as Bicomponent Scaffolds for Tissue Engineering. ACS APPLIED BIO MATERIALS 2024; 7:4593-4601. [PMID: 38914048 DOI: 10.1021/acsabm.4c00469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
Protein-based ultrafine fibrous scaffolds can mimic the native extracellular matrices (ECMs) with regard to the morphology and chemical composition but suffer from poor mechanical and wet stability. As a result, cells cannot get a true three-dimensional (3D) environment as they find in native ECMs. In this study, an epoxide, ethylene glycol diglycidylether (EGDE), with high reactivity to active hydrogen is introduced to gelatin solution, serving as an effective cross-linker. The gelatin/EGDE 3D-ultrafine (∼500 nm in diameter) fibrous composite scaffolds are made by an ultralow-concentration phase separation technique (ULCPS). The effects of the polymer content and modification conditions on the morphology and wet stability of the constructs are investigated. It is revealed that ultrafine fibers with 3D random orientation could be formed at low concentrations (0.01, 0.05, and 0.1 wt %, respectively). The wet stability of the constructs could be effectively improved by introducing EGDE into the gelatin system. The shrinkage is reduced to merely 2.14% after the modification at 120 °C for 2 h and could be maintained for up to 3 days. In order to improve the compression properties, the same technique is utilized with the presence of a poly(lactic acid) (PLA) spacer fabric to produce a bicomponent scaffold. The mechanical property and cell viability of the bicomponent scaffolds are investigated, and it is found that cells could enter deep inside and orient themselves randomly at the central area of the bicomponent scaffold. The modification and design approach presented in this study has the potential to provide various protein-based ultrafine fibrous biomaterials for a variety of biomedical applications.
Collapse
Affiliation(s)
- S M Kamrul Hasan
- Department of Textile Engineering, National Institute of Textile Engineering and Research (NITER), University of Dhaka, Dhaka 1350, Bangladesh
- Shanghai Frontier Science Research Center for Modern Textiles, College of Textiles, Donghua University, Shanghai 201620, China
- Department of Fashion and Textiles, School of Design and Social Context, RMIT University, 25 Dawson Street, Brunswick, Victoria 3054, Australia
| | - Syed Rashedul Islam
- Department of Textile Engineering, National Institute of Textile Engineering and Research (NITER), University of Dhaka, Dhaka 1350, Bangladesh
- Shanghai Frontier Science Research Center for Modern Textiles, College of Textiles, Donghua University, Shanghai 201620, China
- Department of Textile Engineering, Apparel Manufacture and Technology, BGMEA University of Fashion and Technology, Dhaka 1230, Bangladesh
| | - Ismat Zerin
- Department of Textile Engineering, National Institute of Textile Engineering and Research (NITER), University of Dhaka, Dhaka 1350, Bangladesh
| | - Toufique Ahmed
- Department of Textile Engineering, National Institute of Textile Engineering and Research (NITER), University of Dhaka, Dhaka 1350, Bangladesh
| | - Sadikur Rahman
- Department of Textile Engineering, National Institute of Textile Engineering and Research (NITER), University of Dhaka, Dhaka 1350, Bangladesh
| |
Collapse
|
15
|
Liu C, Yang QQ, Zhou YL. Peptides and Wound Healing: From Monomer to Combination. Int J Pept Res Ther 2024; 30:46. [DOI: 10.1007/s10989-024-10627-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/29/2024] [Indexed: 01/02/2025]
|
16
|
Pereira Vasconcelos D, Leite Pereira C, Couto M, Neto E, Ribeiro B, Albuquerque F, Freitas A, Alves CJ, Klinkenberg G, McDonagh BH, Schmid RB, Seitz AM, de Roy L, Ignatius A, Haaparanta A, Muhonen V, Sarmento B, Lamghari M. Nanoenabled Immunomodulatory Scaffolds for Cartilage Tissue Engineering. ADVANCED FUNCTIONAL MATERIALS 2024; 34. [DOI: 10.1002/adfm.202400627] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Indexed: 01/06/2025]
Abstract
AbstractArticular cartilage regeneration is a challenge in tissue engineering. Although diverse materials have been developed for this purpose, cartilage regeneration remains suboptimal. The integration of nanomaterials into 3D network materials holds great potential in the improvement of key mechanical properties, particularly important for osteochondral replacement scaffolds and even to function as carriers for disease‐modifying drugs or other regulatory signals. In this study, a simple yet effective cell‐free nanoenabled Col‐PLA scaffold specially designed to enhance cartilage regeneration and modulate inflammatory response is proposed, by incorporating poly(lactic‐co‐glycolic acid) (PLGA) ibuprofen nanoparticles (NPs) into a collagen/polylactide (Col‐PLA) matrix. The developed nanoenabled scaffold successfully decreases IL‐1β release and leads to primary human chondrocytes survival, ultimately restoring extracellular matrix (ECM) production under inflammatory conditions. The nanoenabled Col‐PLA scaffolds secretome effectively decreases macrophage invasion in vitro, as well as neutrophil infiltration and inflammatory mediators’, namely the complement component C5/C5a, C‐reactive protein, IL‐1β, MMP9, CCL20, and CXCL1/KC production in vivo in a rodent air‐pouch model. Overall, the established nanoenabled scaffold has the potential to support chondrogenesis as well as modulate inflammatory response, overcoming the limitations of traditional tissue engineering strategies.
Collapse
Affiliation(s)
- Daniela Pereira Vasconcelos
- i3S ‐ Instituto de Inovação e Investigação em Saúde INEB ‐ Instituto Nacional de Engenharia Biomédica Universidade do Porto Rua Alfredo Allen, 208 Porto 4200‐135 Portugal
| | - Catarina Leite Pereira
- i3S ‐ Instituto de Inovação e Investigação em Saúde INEB ‐ Instituto Nacional de Engenharia Biomédica Universidade do Porto Rua Alfredo Allen, 208 Porto 4200‐135 Portugal
| | - Marina Couto
- i3S ‐ Instituto de Inovação e Investigação em Saúde INEB ‐ Instituto Nacional de Engenharia Biomédica Universidade do Porto Rua Alfredo Allen, 208 Porto 4200‐135 Portugal
| | - Estrela Neto
- i3S ‐ Instituto de Inovação e Investigação em Saúde INEB ‐ Instituto Nacional de Engenharia Biomédica Universidade do Porto Rua Alfredo Allen, 208 Porto 4200‐135 Portugal
| | - Beatriz Ribeiro
- i3S ‐ Instituto de Inovação e Investigação em Saúde INEB ‐ Instituto Nacional de Engenharia Biomédica Universidade do Porto Rua Alfredo Allen, 208 Porto 4200‐135 Portugal
| | - Filipe Albuquerque
- i3S ‐ Instituto de Inovação e Investigação em Saúde INEB ‐ Instituto Nacional de Engenharia Biomédica Universidade do Porto Rua Alfredo Allen, 208 Porto 4200‐135 Portugal
- School of Medicine and Biomedical Sciences (ICBAS‐UP) Faculdade de Engenharia (FEUP) Universidade do Porto Porto 4050‐313 Portugal
| | - Alexandra Freitas
- i3S ‐ Instituto de Inovação e Investigação em Saúde INEB ‐ Instituto Nacional de Engenharia Biomédica Universidade do Porto Rua Alfredo Allen, 208 Porto 4200‐135 Portugal
- School of Medicine and Biomedical Sciences (ICBAS‐UP) Faculdade de Engenharia (FEUP) Universidade do Porto Porto 4050‐313 Portugal
| | - Cecília J. Alves
- i3S ‐ Instituto de Inovação e Investigação em Saúde INEB ‐ Instituto Nacional de Engenharia Biomédica Universidade do Porto Rua Alfredo Allen, 208 Porto 4200‐135 Portugal
| | - Geir Klinkenberg
- SINTEF Industry Department of Biotechnology and Nanomedicine Trondheim 7034 Norway
| | | | | | - Andreas M. Seitz
- Institute of Orthopedic Research and Biomechanics Center for Trauma Research Ulm University Medical Center Ulm 89081 Ulm Germany
| | - Luisa de Roy
- Institute of Orthopedic Research and Biomechanics Center for Trauma Research Ulm University Medical Center Ulm 89081 Ulm Germany
| | - Anita Ignatius
- Institute of Orthopedic Research and Biomechanics Center for Trauma Research Ulm University Medical Center Ulm 89081 Ulm Germany
| | | | - Virpi Muhonen
- Askel Healthcare Ltd Siltasaarenkatu 8‐10 Helsinki 00530 Finland
| | - Bruno Sarmento
- i3S ‐ Instituto de Inovação e Investigação em Saúde INEB ‐ Instituto Nacional de Engenharia Biomédica Universidade do Porto Rua Alfredo Allen, 208 Porto 4200‐135 Portugal
- Instituto de Investigação e Formação Avançada em Ciências e Tecnologias da Saúde Gandra 4585‐116 Portugal
| | - Meriem Lamghari
- i3S ‐ Instituto de Inovação e Investigação em Saúde INEB ‐ Instituto Nacional de Engenharia Biomédica Universidade do Porto Rua Alfredo Allen, 208 Porto 4200‐135 Portugal
| |
Collapse
|
17
|
Faeed M, Ghiasvand M, Fareghzadeh B, Taghiyar L. Osteochondral organoids: current advances, applications, and upcoming challenges. Stem Cell Res Ther 2024; 15:183. [PMID: 38902814 PMCID: PMC11191177 DOI: 10.1186/s13287-024-03790-5] [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: 02/05/2024] [Accepted: 06/09/2024] [Indexed: 06/22/2024] Open
Abstract
In the realm of studying joint-related diseases, there is a continuous quest for more accurate and representative models. Recently, regenerative medicine and tissue engineering have seen a growing interest in utilizing organoids as powerful tools for studying complex biological systems in vitro. Organoids, three-dimensional structures replicating the architecture and function of organs, provide a unique platform for investigating disease mechanisms, drug responses, and tissue regeneration. The surge in organoid research is fueled by the need for physiologically relevant models to bridge the gap between traditional cell cultures and in vivo studies. Osteochondral organoids have emerged as a promising avenue in this pursuit, offering a better platform to mimic the intricate biological interactions within bone and cartilage. This review explores the significance of osteochondral organoids and the need for their development in advancing our understanding and treatment of bone and cartilage-related diseases. It summarizes osteochondral organoids' insights and research progress, focusing on their composition, materials, cell sources, and cultivation methods, as well as the concept of organoids on chips and application scenarios. Additionally, we address the limitations and challenges these organoids face, emphasizing the necessity for further research to overcome these obstacles and facilitate orthopedic regeneration.
Collapse
Affiliation(s)
- Maryam Faeed
- Cell and Molecular School of Biology, College of Science, University of Tehran, Tehran, Iran
| | - Mahsa Ghiasvand
- Department of Animal Sciences and Marine Biology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran
- Department of Stem Cell and Developmental Biology, Cell Science Research Center, Royan Institute for Stem cell Biology and Technology, ACECR, Tehran, Iran
| | - Bahar Fareghzadeh
- Department of Biomedical Engineering, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Leila Taghiyar
- Department of Stem Cell and Developmental Biology, Cell Science Research Center, Royan Institute for Stem cell Biology and Technology, ACECR, Tehran, Iran.
- Advanced Therapy Medicinal Product Technology Development Center (ATMP-TDC), Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| |
Collapse
|
18
|
Primorac D, Molnar V, Tsoukas D, Uzieliene I, Tremolada C, Brlek P, Klarić E, Vidović D, Zekušić M, Pachaleva J, Bernotiene E, Wilson A, Mobasheri A. Tissue engineering and future directions in regenerative medicine for knee cartilage repair: a comprehensive review. Croat Med J 2024; 65:268-287. [PMID: 38868973 PMCID: PMC11157252 DOI: 10.3325/cmj.2024.65.268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 05/26/2024] [Indexed: 01/06/2025] Open
Abstract
This review evaluates the current landscape and future directions of regenerative medicine for knee cartilage repair, with a particular focus on tissue engineering strategies. In this context, scaffold-based approaches have emerged as promising solutions for cartilage regeneration. Synthetic scaffolds, while offering superior mechanical properties, often lack the biological cues necessary for effective tissue integration. Natural scaffolds, though biocompatible and biodegradable, frequently suffer from inadequate mechanical strength. Hybrid scaffolds, combining elements of both synthetic and natural materials, present a balanced approach, enhancing both mechanical support and biological functionality. Advances in decellularized extracellular matrix scaffolds have shown potential in promoting cell infiltration and integration with native tissues. Additionally, bioprinting technologies have enabled the creation of complex, bioactive scaffolds that closely mimic the zonal organization of native cartilage, providing an optimal environment for cell growth and differentiation. The review also explores the potential of gene therapy and gene editing techniques, including CRISPR-Cas9, to enhance cartilage repair by targeting specific genetic pathways involved in tissue regeneration. The integration of these advanced therapies with tissue engineering approaches holds promise for developing personalized and durable treatments for knee cartilage injuries and osteoarthritis. In conclusion, this review underscores the importance of continued multidisciplinary collaboration to advance these innovative therapies from bench to bedside and improve outcomes for patients with knee cartilage damage.
Collapse
Affiliation(s)
- Dragan Primorac
- Dragan Primorac, Poliklinika Sv. Katarina, Branimirova 71E, 10000 Zagreb, Croatia,
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Wojciechowski C, Wasyłeczko M, Lewińska D, Chwojnowski A. A Comprehensive Review of Hollow-Fiber Membrane Fabrication Methods across Biomedical, Biotechnological, and Environmental Domains. Molecules 2024; 29:2637. [PMID: 38893513 PMCID: PMC11174095 DOI: 10.3390/molecules29112637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 05/29/2024] [Accepted: 05/30/2024] [Indexed: 06/21/2024] Open
Abstract
This work presents methods of obtaining polymeric hollow-fiber membranes produced via the dry-wet phase inversion method that were published in renowned specialized membrane publications in the years 2010-2020. Obtaining hollow-fiber membranes, unlike flat membranes, requires the use of a special installation for their production, the most important component of which is the hollow fiber forming spinneret. This method is most often used in obtaining membranes made of polysulfone, polyethersulfone, polyurethane, cellulose acetate, and its derivatives. Many factors affect the properties of the membranes obtained. By changing the parameters of the spinning process, we change the thickness of the membranes' walls and the diameter of the hollow fibers, which causes changes in the membranes' structure and, as a consequence, changes in their transport/separation parameters. The type of bore fluid affects the porosity of the inner epidermal layer or causes its atrophy. Porogenic compounds such as polyvinylpyrrolidones and polyethylene glycols and other substances that additionally increase the membrane porosity are often added to the polymer solution. Another example is a blend of two- or multi-component membranes and dual-layer membranes that are obtained using a three-nozzle spinneret. In dual-layer membranes, one layer is the membrane scaffolding, and the other is the separation layer. Also, the temperature during the process, the humidity, and the composition of the solution in the coagulating bath have impact on the parameters of the membranes obtained.
Collapse
Affiliation(s)
- Cezary Wojciechowski
- Nalecz Institute of Biocybernetic and Biomedical Engineering, Polish Academy of Sciences, Trojdena 4 Str., 02-109 Warsaw, Poland; (M.W.); (D.L.); (A.C.)
| | | | | | | |
Collapse
|
20
|
Tamo AK, Djouonkep LDW, Selabi NBS. 3D Printing of Polysaccharide-Based Hydrogel Scaffolds for Tissue Engineering Applications: A Review. Int J Biol Macromol 2024; 270:132123. [PMID: 38761909 DOI: 10.1016/j.ijbiomac.2024.132123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 05/02/2024] [Accepted: 05/04/2024] [Indexed: 05/20/2024]
Abstract
In tissue engineering, 3D printing represents a versatile technology employing inks to construct three-dimensional living structures, mimicking natural biological systems. This technology efficiently translates digital blueprints into highly reproducible 3D objects. Recent advances have expanded 3D printing applications, allowing for the fabrication of diverse anatomical components, including engineered functional tissues and organs. The development of printable inks, which incorporate macromolecules, enzymes, cells, and growth factors, is advancing with the aim of restoring damaged tissues and organs. Polysaccharides, recognized for their intrinsic resemblance to components of the extracellular matrix have garnered significant attention in the field of tissue engineering. This review explores diverse 3D printing techniques, outlining distinctive features that should characterize scaffolds used as ideal matrices in tissue engineering. A detailed investigation into the properties and roles of polysaccharides in tissue engineering is highlighted. The review also culminates in a profound exploration of 3D polysaccharide-based hydrogel applications, focusing on recent breakthroughs in regenerating different tissues such as skin, bone, cartilage, heart, nerve, vasculature, and skeletal muscle. It further addresses challenges and prospective directions in 3D printing hydrogels based on polysaccharides, paving the way for innovative research to fabricate functional tissues, enhancing patient care, and improving quality of life.
Collapse
Affiliation(s)
- Arnaud Kamdem Tamo
- Institute of Microsystems Engineering IMTEK, University of Freiburg, 79110 Freiburg, Germany; Freiburg Center for Interactive Materials and Bioinspired Technologies FIT, University of Freiburg, 79110 Freiburg, Germany; Freiburg Materials Research Center FMF, University of Freiburg, 79104 Freiburg, Germany; Ingénierie des Matériaux Polymères (IMP), Université Claude Bernard Lyon 1, INSA de Lyon, Université Jean Monnet, CNRS, UMR 5223, 69622 Villeurbanne CEDEX, France.
| | - Lesly Dasilva Wandji Djouonkep
- College of Petroleum Engineering, Yangtze University, Wuhan 430100, China; Key Laboratory of Drilling and Production Engineering for Oil and Gas, Wuhan 430100, China
| | - Naomie Beolle Songwe Selabi
- Institute of Advanced Materials and Nanotechnology, Wuhan University of Science and Technology, Wuhan 430081, China
| |
Collapse
|
21
|
Farzamfar S, Garcia LM, Rahmani M, Bolduc S. Navigating the Immunological Crossroads: Mesenchymal Stem/Stromal Cells as Architects of Inflammatory Harmony in Tissue-Engineered Constructs. Bioengineering (Basel) 2024; 11:494. [PMID: 38790361 PMCID: PMC11118848 DOI: 10.3390/bioengineering11050494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 04/26/2024] [Accepted: 05/13/2024] [Indexed: 05/26/2024] Open
Abstract
In the dynamic landscape of tissue engineering, the integration of tissue-engineered constructs (TECs) faces a dual challenge-initiating beneficial inflammation for regeneration while avoiding the perils of prolonged immune activation. As TECs encounter the immediate reaction of the immune system upon implantation, the unique immunomodulatory properties of mesenchymal stem/stromal cells (MSCs) emerge as key navigators. Harnessing the paracrine effects of MSCs, researchers aim to craft a localized microenvironment that not only enhances TEC integration but also holds therapeutic promise for inflammatory-driven pathologies. This review unravels the latest advancements, applications, obstacles, and future prospects surrounding the strategic alliance between MSCs and TECs, shedding light on the immunological symphony that guides the course of regenerative medicine.
Collapse
Affiliation(s)
- Saeed Farzamfar
- Centre de Recherche en Organogénèse Expérimentale/LOEX, Regenerative Medicine Division, CHU de Québec-Université Laval Research Center, Québec, QC G1V 4G2, Canada; (S.F.); (M.R.)
| | - Luciana Melo Garcia
- Department of Medicine, Université Laval, Québec, QC G1V 0A6, Canada;
- Hematology-Oncology Service, CHU de Québec—Université Laval, Québec, QC G1V 0A6, Canada
| | - Mahya Rahmani
- Centre de Recherche en Organogénèse Expérimentale/LOEX, Regenerative Medicine Division, CHU de Québec-Université Laval Research Center, Québec, QC G1V 4G2, Canada; (S.F.); (M.R.)
| | - Stephane Bolduc
- Centre de Recherche en Organogénèse Expérimentale/LOEX, Regenerative Medicine Division, CHU de Québec-Université Laval Research Center, Québec, QC G1V 4G2, Canada; (S.F.); (M.R.)
- Department of Surgery, Faculty of Medicine, Université Laval, Québec, QC G1V 0A6, Canada
| |
Collapse
|
22
|
Damiati LA, El Soury M. Bone-nerve crosstalk: a new state for neuralizing bone tissue engineering-A mini review. Front Med (Lausanne) 2024; 11:1386683. [PMID: 38690172 PMCID: PMC11059066 DOI: 10.3389/fmed.2024.1386683] [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: 02/15/2024] [Accepted: 03/18/2024] [Indexed: 05/02/2024] Open
Abstract
Neuro bone tissue engineering is a multidisciplinary field that combines both principles of neurobiology and bone tissue engineering to develop innovative strategies for repairing and regenerating injured bone tissues. Despite the fact that regeneration and development are considered two distinct biological processes, yet regeneration can be considered the reactivation of development in later life stages to restore missing tissues. It is noteworthy that the regeneration capabilities are distinct and vary from one organism to another (teleost fishes, hydra, humans), or even in the same organism can vary dependent on the injured tissue itself (Human central nervous system vs. peripheral nervous system). The skeletal tissue is highly innervated, peripheral nervous system plays a role in conveying the signals and connecting the central nervous system with the peripheral organs, moreover it has been shown that they play an important role in tissue regeneration. Their regeneration role is conveyed by the different cells' resident in it and in its endoneurium (fibroblasts, microphages, vasculature associated cells, and Schwann cells) these cells secrete various growth factors (NGF, BDNF, GDNF, NT-3, and bFGF) that contribute to the regenerative phenotype. The peripheral nervous system and central nervous system synchronize together in regulating bone homeostasis and regeneration through neurogenic factors and neural circuits. Receptors of important central nervous system peptides such as Serotonin, Leptin, Semaphorins, and BDNF are expressed in bone tissue playing a role in bone homeostasis, metabolism and regeneration. This review will highlight the crosstalk between peripheral nerves and bone in the developmental stages as well as in regeneration and different neuro-bone tissue engineering strategies for repairing severe bone injuries.
Collapse
Affiliation(s)
- Laila A. Damiati
- Department of Biological Sciences, College of Science, University of Jeddah, Jeddah, Saudi Arabia
| | - Marwa El Soury
- Department of Clinical and Biological Sciences, University of Torino, Torino, Italy
- Neuroscience Institute Cavalieri Ottolenghi (NICO), University of Torino, Orbassano, Italy
| |
Collapse
|
23
|
Wasyłeczko M, Wojciechowski C, Chwojnowski A. Polyethersulfone Polymer for Biomedical Applications and Biotechnology. Int J Mol Sci 2024; 25:4233. [PMID: 38673817 PMCID: PMC11049998 DOI: 10.3390/ijms25084233] [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: 03/07/2024] [Revised: 04/03/2024] [Accepted: 04/09/2024] [Indexed: 04/28/2024] Open
Abstract
Polymers stand out as promising materials extensively employed in biomedicine and biotechnology. Their versatile applications owe much to the field of tissue engineering, which seamlessly integrates materials engineering with medical science. In medicine, biomaterials serve as prototypes for organ development and as implants or scaffolds to facilitate body regeneration. With the growing demand for innovative solutions, synthetic and hybrid polymer materials, such as polyethersulfone, are gaining traction. This article offers a concise characterization of polyethersulfone followed by an exploration of its diverse applications in medical and biotechnological realms. It concludes by summarizing the significant roles of polyethersulfone in advancing both medicine and biotechnology, as outlined in the accompanying table.
Collapse
Affiliation(s)
- Monika Wasyłeczko
- Nalecz Institute of Biocybernetics and Biomedical Engineering, Polish Academy of Sciences, Ksiecia Trojdena 4, 02-109 Warsaw, Poland; (C.W.); (A.C.)
| | | | | |
Collapse
|
24
|
Mamidi N, Ijadi F, Norahan MH. Leveraging the Recent Advancements in GelMA Scaffolds for Bone Tissue Engineering: An Assessment of Challenges and Opportunities. Biomacromolecules 2024; 25:2075-2113. [PMID: 37406611 DOI: 10.1021/acs.biomac.3c00279] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/07/2023]
Abstract
The field of bone tissue engineering has seen significant advancements in recent years. Each year, over two million bone transplants are performed globally, and conventional treatments, such as bone grafts and metallic implants, have their limitations. Tissue engineering offers a new level of treatment, allowing for the creation of living tissue within a biomaterial framework. Recent advances in biomaterials have provided innovative approaches to rebuilding bone tissue function after damage. Among them, gelatin methacryloyl (GelMA) hydrogel is emerging as a promising biomaterial for supporting cell proliferation and tissue regeneration, and GelMA has exhibited exceptional physicochemical and biological properties, making it a viable option for clinical translation. Various methods and classes of additives have been used in the application of GelMA for bone regeneration, with the incorporation of nanofillers or other polymers enhancing its resilience and functional performance. Despite promising results, the fabrication of complex structures that mimic the bone architecture and the provision of balanced physical properties for both cell and vasculature growth and proper stiffness for load bearing remain as challenges. In terms of utilizing osteogenic additives, the priority should be on versatile components that promote angiogenesis and osteogenesis while reinforcing the structure for bone tissue engineering applications. This review focuses on recent efforts and advantages of GelMA-based composite biomaterials for bone tissue engineering, covering the literature from the last five years.
Collapse
Affiliation(s)
- Narsimha Mamidi
- Department of Chemistry and Nanotechnology, School of Engineering and Science, Tecnológico de Monterrey, Monterrey, Nuevo León 64849, México
- Wisconsin Center for NanoBioSystems, School of Pharmacy, University of Wisconsin, Madison, Wisconsin 53705, United States
| | - Fatemeh Ijadi
- Department of Chemistry and Nanotechnology, School of Engineering and Science, Tecnológico de Monterrey, Monterrey, Nuevo León 64849, México
| | - Mohammad Hadi Norahan
- Centro de Biotecnología-FEMSA, Tecnológico de Monterrey, Monterrey, Nuevo León 64849, México
| |
Collapse
|
25
|
Iravani K, Mousavi S, Owji SM, Sani M, Owji SH. Effect of amniotic membrane/collagen scaffolds on laryngeal cartilage repair. Laryngoscope Investig Otolaryngol 2024; 9:e1222. [PMID: 38362193 PMCID: PMC10866587 DOI: 10.1002/lio2.1222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Accepted: 01/16/2024] [Indexed: 02/17/2024] Open
Abstract
Objectives Laryngeal cartilage defects are a major problem that greatly impacts structural integrity and function. Cartilage repair is also a challenging issue. This study evaluated the efficacy of a collagen scaffold enveloped by amniotic membrane (AM/C) on laryngeal cartilage repair. Study Design Experimental animal study. Methods Fourteen Dutch rabbits were enrolled in the study. A 5 mm cartilage defect was created in the right and left thyroid lamina. The animals were divided into two groups randomly. Group 1 collagen scaffolds and group 2 AM/C were applied to the right side defects. Left side defects were not repaired, serving as control. Histologic evaluation was done 45 and 90 days following collagen and AM/C application with criteria of tissue and cell morphology, lacuna formation, vascularization, and inflammation. Results Significant improvement in cartilage repair was observed in the AM/C side compared to the control side in all histologic criteria after 45 days (p<.05). After 90 days, cartilage repair improved in cell morphology, lacuna formation, and inflammation significantly (p<.05). Conclusion The combination of amniotic membrane and collagen scaffolds provides a promising treatment modality for improving the repair of laryngeal cartilage defects. Level of Evidence NA.
Collapse
Affiliation(s)
- Kamyar Iravani
- Otolaryngology Research Center, Department of OtolaryngologyShiraz University of Medical SciencesShirazIran
| | - Simin Mousavi
- Otolaryngology Research Center, Department of OtolaryngologyShiraz University of Medical SciencesShirazIran
| | - Seyed Mohammad Owji
- Department of PathologySchool of Medicine, Shiraz University of Medical SciencesShirazIran
| | - Mahsa Sani
- Shiraz Institute for Stem Cell and Regenerative Medicine, Shiraz University of Medical SciencesShirazIran
| | - Seyed Hossein Owji
- Otolaryngology Research Center, Department of OtolaryngologyShiraz University of Medical SciencesShirazIran
| |
Collapse
|
26
|
Kolar M, Veber M, Girandon L, Drobnič M. Biomaterials augmented with filtered bone marrow aspirate for the treatment of talar osteochondral lesions. A comparison of clinical and cellular parameters. J Orthop Surg (Hong Kong) 2024; 32:10225536231219970. [PMID: 38214308 DOI: 10.1177/10225536231219970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2024] Open
Abstract
BACKGROUND Biomaterials augmented with Bone Marrow Aspirate Concentrate (BMAC) are becoming increasingly utilized in the cartilage treatment. However, the potential role of cellular parameters in the intraoperatively applied BMAC have yet to be elucidated. PURPOSE (A) To evaluate clinical outcomes and safety of a combined single-step approach with scaffolds (fibrin glues, collagen gels, collagen-hydroxyapatite membrane) and filtered Bone Marrow Aspirate (fBMA) for the treatment of osteochondral lesions of the talus (OLTs). (B) To identify significant factors for postoperative improvements, considering cellular parameters as potential predictors. METHODS All the patients operated on due to OLTs by the combination above were selected from the hospital registry database (35 pts, years 16-55, and minimally 1 year follow-up). Treatment outcomes were followed clinically with Patient-reported outcome measures (PROMs), and by pursuing serious adverse events (SAE) and graft failures (GF). Cellular parameters of the injected fBMA were determined. Pre- and postoperative PROMs values were compared to evaluate postoperative improvements. Multivariable regression models were applied to identify potential factors (demographics, medical history, joint and lesion characteristics, scaffold type, surgical and cellular parameters) that predict the treatment outcomes. RESULTS At the mean follow-up of 32.2 (12.5) months, all Foot and Ankle Outcome Score (FAOS) and European Quality of Life in Five Dimensions Three-Level (EQ-5D-3 L) values improved significantly. 4 (11%) SAE (3 arthrofibrosis, one hardware removal), and 3 (9%) GF occurred. Female gender and concomitant procedures were the main negative predictors for postoperative outcomes. The number of fibroblast colony forming units (CFU-F) or their proportion among total nucleated cells (CFU-F/TNC) were positively correlated with the improvements of some PROMs. CONCLUSIONS Scaffolds augmented with fBMA proved as an adequate and safe approach for OLTs treatment. Cellular parameters seem to influence the treatment outcomes, thus further attention should be given to the intraoperatively applied products. LEVEL OF EVIDENCE Level IV.
Collapse
Affiliation(s)
- Matic Kolar
- Department of Orthopaedic Surgery, University Medical Centre Ljubljana, Ljubljana, Slovenia
- Chair of Orthopaedics, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | | | | | - Matej Drobnič
- Department of Orthopaedic Surgery, University Medical Centre Ljubljana, Ljubljana, Slovenia
- Chair of Orthopaedics, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
27
|
Jeyaraman M, Nallakumarasamy A, Jeyaraman N, Ramasubramanian S. Tissue engineering in chondral defect. COMPUTATIONAL BIOLOGY FOR STEM CELL RESEARCH 2024:361-378. [DOI: 10.1016/b978-0-443-13222-3.00033-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
28
|
Gupta P, Sharma S, Jabin S, Jadoun S. Chitosan nanocomposite for tissue engineering and regenerative medicine: A review. Int J Biol Macromol 2024; 254:127660. [PMID: 37907176 DOI: 10.1016/j.ijbiomac.2023.127660] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 10/18/2023] [Accepted: 10/23/2023] [Indexed: 11/02/2023]
Abstract
Regenerative medicine and tissue engineering have emerged as a multidisciplinary promising field in the quest to address the limitations of traditional medical approaches. One of the key aspects of these fields is the development of such types of biomaterials that can mimic the extracellular matrix and provide a conducive environment for tissue regeneration. In this regard, chitosan has played a vital role which is a naturally derived linear bi-poly-aminosaccharide, and has gained significant attention due to its biocompatibility and unique properties. Chitosan possesses many unique physicochemical properties, making it a significant polysaccharide for different applications such as agriculture, nutraceutical, biomedical, food, nutraceutical, packaging, etc. as well as significant material for developing next-generation hydrogel and bio-scaffolds for regenerative medicinal applications. Moreover, chitosan can be easily modified to incorporate desirable properties, such as improved mechanical strength, enhanced biodegradability, and controlled release of bioactive molecules. Blending chitosan with other polymers or incorporating nanoparticles into its matrix further expands its potential in tissue engineering applications. This review summarizes the most recent studies of the last 10 years based on chitosan, blends, and nanocomposites and their application in bone tissue engineering, hard tissue engineering, dental implants, dental tissue engineering, dental fillers, and cartilage tissue engineering.
Collapse
Affiliation(s)
- Priti Gupta
- Department of Chemistry, Manav Rachna University, Faridabad, Haryana 121001, India.
| | - Shilpa Sharma
- Department of Chemistry, Manav Rachna University, Faridabad, Haryana 121001, India.
| | - Shagufta Jabin
- Department of Chemistry, Faculty of Engineering, Manav Rachna International Institute of Research & Studies, Faridabad, India.
| | - Sapana Jadoun
- Departamento de Química, Facultad de Ciencias, Universidad de Tarapacá, Avda. General Velásquez, 1775 Arica, Chile.
| |
Collapse
|
29
|
Saghati S, Avci ÇB, Hassani A, Nazifkerdar S, Amini H, Saghebasl S, Mahdipour M, Banimohamad-Shotorbani B, Namjoo AR, Abrbekoh FN, Rahbarghazi R, Nasrabadi HT, Khoshfetrat AB. Phenolated alginate hydrogel induced osteogenic properties of mesenchymal stem cells via Wnt signaling pathway. Int J Biol Macromol 2023; 253:127209. [PMID: 37804896 DOI: 10.1016/j.ijbiomac.2023.127209] [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: 08/20/2022] [Revised: 09/06/2023] [Accepted: 10/01/2023] [Indexed: 10/09/2023]
Abstract
Osteogenic properties of phenolated alginate (1.2 %) hydrogel containing collagen (0.5 %)/nano-hydroxyapatite (1 %) were studied on human mesenchymal stem cells in vitro. The phenolation rate and physical properties of the hydrogel were assessed using nuclear magnetic resonance (NMR), Fourier-transform infrared spectroscopy (FTIR), Scanning electron microscope (SEM), swelling ratio, gelation time, mechanical assay, and degradation rate. The viability of encapsulated cells was monitored on days 7, 14, and 21 using an MTT assay. Osteoblast differentiation was studied using western blotting, and real-time PCR. Using PCR array analysis, the role of the Wnt signaling pathway was also investigated. Data showed that the combination of alginate/collagen/nanohydroxyapatite yielded proper mechanical features. The addition of nanohydroxyapatite, and collagen reduced degradation, swelling rate coincided with increased stiffness. Elasticity and pore size were also diminished. NMR and FTIR revealed suitable incorporation of collagen and nanohydroxyapatite in the structure of alginate. Real-time PCR analysis and western blotting indicated the expression of osteoblast-related genes such as Runx2 and osteocalcin. PCR array revealed the induction of numerous genes related to Wnt signaling pathways during the maturation of human stem cells toward osteoblast-like cells. In vivo data indicated that transplantation of phenolated alginate/collagen/nanohydroxyapatite hydrogel led to enhanced de novo bone formation in rats with critical-sized calvarial defects. Phenolated alginate hydrogel can promote the osteogenic capacity of human amniotic membrane mesenchymal stem cells in the presence of nanohydroxyapatite and collagen via engaging the Wnt signaling pathway.
Collapse
Affiliation(s)
- Sepideh Saghati
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Tissue Engineering, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran; Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Çığır Biray Avci
- Department of Medical Biology, Faculty of Medicine, Ege University, Izmir, Turkey
| | - Ayla Hassani
- Chemical Engineering Faculty, Sahand University of Technology, Tabriz 51335-1996, Iran; Stem Cell and Tissue Engineering Research Laboratory, Sahand University of Technology, Tabriz 51335-1996, Iran
| | - Sajed Nazifkerdar
- Chemical Engineering Faculty, Sahand University of Technology, Tabriz 51335-1996, Iran; Stem Cell and Tissue Engineering Research Laboratory, Sahand University of Technology, Tabriz 51335-1996, Iran
| | - Hassan Amini
- Department of General and Vascular Surgery, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Solmaz Saghebasl
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mahdi Mahdipour
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behnaz Banimohamad-Shotorbani
- Department of Tissue Engineering, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Atieh Rezaei Namjoo
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Reza Rahbarghazi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Hamid Tayefi Nasrabadi
- Department of Tissue Engineering, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Ali Baradar Khoshfetrat
- Department of Medical Biology, Faculty of Medicine, Ege University, Izmir, Turkey; Chemical Engineering Faculty, Sahand University of Technology, Tabriz 51335-1996, Iran.
| |
Collapse
|
30
|
Kalvand E, Bakhshandeh H, Nadri S, Habibizadeh M, Rostamizadeh K. Poly-ε-caprolactone (PCL)/poly-l-lactic acid (PLLA) nanofibers loaded by nanoparticles-containing TGF-β1 with linearly arranged transforming structure as a scaffold in cartilage tissue engineering. J Biomed Mater Res A 2023; 111:1838-1849. [PMID: 37395312 DOI: 10.1002/jbm.a.37574] [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: 02/18/2023] [Revised: 04/24/2023] [Accepted: 05/16/2023] [Indexed: 07/04/2023]
Abstract
This study aimed to present a novel three-dimensional nanocomposite scaffold using poly-ε-caprolactone (PCL), containing transforming growth factor-beta 1 (TGF-β1)-loaded chitosan-dextran nanoparticles and poly-l-lactic acid (PLLA), to make use of nanofibers and nanoparticles simultaneously. The electrospinning method fabricated a bead-free semi-aligned nanofiber composed of PLLA, PCL, and chitosan-dextran nanoparticles containing TGF-β1. A biomimetic scaffold was constructed with the desired mechanical properties, high hydrophilicity, and high porosity. Transmission electron microscopy findings showed a linear arrangement of nanoparticles along the core of fibers. Based on the results, burst release was not observed. The maximum release was achieved within 4 days, and sustained release was up to 21 days. The qRT-PCR results indicated an increase in the expression of aggrecan and collagen type Ι genes compared to the tissue culture polystyrene group. The results indicated the importance of topography and the sustained release of TGF-β1 from bifunctional scaffolds in directing the stem cell fate in cartilage tissue engineering.
Collapse
Affiliation(s)
- Elham Kalvand
- Department of Pharmaceutical Biomaterials, School of Pharmacy, Zanjan University of Medical Sciences, Zanjan, Iran
- Department of Nanobiotechnology, Pasteur Institute of Tehran, Tehran, Iran
- Department of Nanotechnology and Tissue Engineering, Stem Cell Technology Research of Tehran, Tehran, Iran
| | - Haleh Bakhshandeh
- Department of Nanobiotechnology, Pasteur Institute of Tehran, Tehran, Iran
- New Technologies Research Group, Department of Nanobiotechnology, Pasteur Institute of Iran, Tehran, Iran
| | - Samad Nadri
- Zanjan Pharmaceutical Nanotechnology Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
- Department of Medical Nanotechnology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Mina Habibizadeh
- Regenerative Medicine Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Kobra Rostamizadeh
- Zanjan Pharmaceutical Nanotechnology Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
- Pharmaceutical Biomaterials Department, School of Pharmacy, Zanjan University of Medical Sciences, Zanjan, Iran
| |
Collapse
|
31
|
Reina-Mahecha A, Beers MJ, van der Veen HC, Zuhorn IS, van Kooten TG, Sharma PK. A Review of the Role of Bioreactors for iPSCs-Based Tissue-Engineered Articular Cartilage. Tissue Eng Regen Med 2023; 20:1041-1052. [PMID: 37861960 PMCID: PMC10645985 DOI: 10.1007/s13770-023-00573-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 07/04/2023] [Accepted: 07/05/2023] [Indexed: 10/21/2023] Open
Abstract
BACKGROUND Osteoarthritis (OA) is the most common degenerative joint disease without an ultimate treatment. In a search for novel approaches, tissue engineering (TE) has shown great potential to be an effective way for hyaline cartilage regeneration and repair in advanced stages of OA. Recently, induced pluripotent stem cells (iPSCs) have been appointed to be essential stem cells for degenerative disease treatment because they allow a personalized medicine approach. For clinical translation, bioreactors in combination with iPSCs-engineerd cartilage could match patients needs, serve as platform for large-scale patient specific cartilage production, and be a tool for patient OA modelling and drug screening. Furthermore, to minimize in vivo experiments and improve cell differentiation and cartilage extracellular matrix (ECM) deposition, TE combines existing approaches with bioreactors. METHODS This review summarizes the current understanding of bioreactors and the necessary parameters when they are intended for cartilage TE, focusing on the potential use of iPSCs. RESULTS Bioreactors intended for cartilage TE must resemble the joint cavity niche. However, recreating human synovial joints is not trivial because the interactions between various stimuli are not entirely understood. CONCLUSION The use of mechanical and electrical stimulation to differentiate iPSCs, and maintain and test chondrocytes are key stimuli influencing hyaline cartilage homeostasis. Incorporating these stimuli to bioreactors can positively impact cartilage TE approaches and their possibility for posterior translation into the clinics.
Collapse
Affiliation(s)
- Alejandro Reina-Mahecha
- Department of Biomedical Engineering, University of Groningen, University Medical Center Groningen, FB40, Antonius Deusinglaan -1, 9713AV, Groningen, The Netherlands
| | - Martine J Beers
- Department of Orthopedics, University Medical Center Groningen, Groningen, The Netherlands
| | - Hugo C van der Veen
- Department of Orthopedics, University Medical Center Groningen, Groningen, The Netherlands
| | - Inge S Zuhorn
- Department of Biomedical Engineering, University of Groningen, University Medical Center Groningen, FB40, Antonius Deusinglaan -1, 9713AV, Groningen, The Netherlands
| | - Theo G van Kooten
- Department of Biomedical Engineering, University of Groningen, University Medical Center Groningen, FB40, Antonius Deusinglaan -1, 9713AV, Groningen, The Netherlands
| | - Prashant K Sharma
- Department of Biomedical Engineering, University of Groningen, University Medical Center Groningen, FB40, Antonius Deusinglaan -1, 9713AV, Groningen, The Netherlands.
| |
Collapse
|
32
|
Thomas V, Mercuri J. In vitro and in vivo efficacy of naturally derived scaffolds for cartilage repair and regeneration. Acta Biomater 2023; 171:1-18. [PMID: 37708926 DOI: 10.1016/j.actbio.2023.09.008] [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: 05/26/2023] [Revised: 08/13/2023] [Accepted: 09/07/2023] [Indexed: 09/16/2023]
Abstract
Intrinsically present bioactive cues allow naturally derived materials to mimic important characteristics of cartilage while also facilitating cellular recruitment, infiltration, and differentiation. Such traits are often what tissue engineers desire when they fabricate scaffolds, and yet, literature from the past decade is replete with examples of how most natural constructs with native biomolecules have only offered sub-optimal results in the treatment of cartilage defects. This paper provides an in-depth investigation of the performance of such scaffolds through a review of a collection of natural materials that have been used so far in repairing/regenerating articular cartilage. Although in vivo and clinical studies are the best indicators of scaffold efficacy, it was, however, observed that a large number of natural constructs had very promising scaffold characteristics to begin with, and would often show good in vitro/in vivo results. Finally, an examination of the biochemistry and biomechanics of repair tissues in studies that reported positive outcomes showed that these attributes often approached target cartilage values. The paper concludes with an outline of current trends as well as future directions for the field. STATEMENT OF SIGNIFICANCE: This review offers an exclusive focus on natural scaffold materials for cartilage repair and regeneration and provides a quantitative and qualitative analysis of their performance under a variety of in vitro and in vivo conditions. Readers can learn about environments where natural scaffolds have had the most success and tailor strategies to optimize their own work. Furthermore, given how the glycosaminoglycan (GAG) to hydroxyproline (HYP) ratio and moduli are fundamental attributes of hyaline cartilage, this paper adds to the body of knowledge by exploring how these characteristics reflect in preclinical outcomes. Such perspectives can greatly aid researchers better utilize natural materials for Cartilage Tissue Engineering (CTE).
Collapse
Affiliation(s)
- Vishal Thomas
- The Laboratory of Orthopaedic Tissue Regeneration & Orthobiologics, Department of Bioengineering, 401-5 Rhodes Engineering Research Center, Clemson, SC 29631, USA
| | - Jeremy Mercuri
- The Laboratory of Orthopaedic Tissue Regeneration & Orthobiologics, Department of Bioengineering, 401-5 Rhodes Engineering Research Center, Clemson, SC 29631, USA.
| |
Collapse
|
33
|
Kacprzak B, Rosińska K. Rehabilitation of Soccer Players' Knee Injuries: Cartilage Reconstruction, Anterior Cruciate Ligament Surgery, and Intensive Recovery-A Pilot Study. J Clin Med 2023; 12:6893. [PMID: 37959358 PMCID: PMC10650160 DOI: 10.3390/jcm12216893] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 10/09/2023] [Accepted: 10/30/2023] [Indexed: 11/15/2023] Open
Abstract
Knee injuries, particularly anterior cruciate ligament (ACL) damage and cartilage defects, are highly prevalent among athletes and affect their sports performance and long-term joint function. The purpose of this research was to evaluate the effectiveness of a comprehensive combination therapy approach for individuals with ACL and cartilage injuries. Twelve professional soccer players aged 18 to 30 years underwent bone-tendon-bone ACL reconstruction, microfracture cartilage repair surgery, and hyaluronic acid scaffold treatment. Early postoperative rehabilitation included immediate supervised physiotherapy and complete weight bearing. Follow-up assessments involved clinical evaluations, functional joint assessments, and magnetic resonance imaging (MRI) scans to measure cartilage defect repair and symptom alleviation. The results showed that patients resumed pain-free activities within 3-4 weeks and returned to their pre-injury level within 4.5 months. MRI demonstrated the absence of inflammatory reactions, repair of marrow edema, and the emergence of new cartilage. Six months and one year after surgery, the Knee Injury and Osteoarthritis Outcome Score (KOOS) and the Short Form (36) Health Survey (SF-36) questionnaire results demonstrated considerable improvement in patients' health condition and quality of life. Overall, the study suggests that the combination of Hyalofast membranes, microfracture surgery, tissue adhesive, and intensive postoperative physical therapy may be a potential alternative to commonly used treatments for patients with ACL rupture, allowing them to recover efficiently and return to sports activities.
Collapse
Affiliation(s)
- Bartłomiej Kacprzak
- Orto Med Sport Łódź, 28 Pułku Strzelców Kaniowskich 45, 90-640 Łódź, Poland;
| | - Karolina Rosińska
- Wolf Project Studio Krzysztof Król, ul. Gdańska 79/D01, 90-613 Łódź, Poland
| |
Collapse
|
34
|
Zavala G, Viafara-García SM, Novoa J, Hidalgo C, Contardo I, Díaz-Calderón P, Alejandro González-Arriagada W, Khoury M, Acevedo JP. An advanced biphasic porous and injectable scaffold displays a fine balance between mechanical strength and remodeling capabilities essential for cartilage regeneration. Biomater Sci 2023; 11:6801-6822. [PMID: 37622217 DOI: 10.1039/d3bm00703k] [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: 08/26/2023]
Abstract
An important challenge in tissue engineering is the regeneration of functional articular cartilage (AC). In the field, biomimetic hydrogels are being extensively studied as scaffolds that recapitulate microenvironmental features or as mechanical supports for transplanted cells. New advanced hydrogel formulations based on salmon methacrylate gelatin (sGelMA), a cold-adapted biomaterial, are presented in this work. The psychrophilic nature of this biomaterial provides rheological advantages allowing the fabrication of scaffolds with high concentrations of the biopolymer and high mechanical strength, suitable for formulating injectable hydrogels with high mechanical strength for cartilage regeneration. However, highly intricate cell-laden scaffolds derived from highly concentrated sGelMA solutions could be deleterious for cells and scaffold remodeling. On this account, the current study proposes the use of sGelMA supplemented with a mesophilic sacrificial porogenic component. The cytocompatibility of different sGelMA-based formulations is tested through the encapsulation of osteoarthritic chondrocytes (OACs) and stimulated to synthesize extracellular matrix (ECM) components in vitro and in vivo. The sGelMA-derived scaffolds reach high levels of stiffness, and the inclusion of porogens impacts positively the scaffold degradability and molecular diffusion, improved fitness of OACs, increased the expression of cartilage-related genes, increased glycosaminoglycan (GAG) synthesis, and improved remodeling toward cartilage-like tissues. Altogether, these data support the use of sGelMA solutions in combination with mammalian solid gelatin beads for highly injectable formulations for cartilage regeneration, strengthening the importance of the balance between mechanical properties and remodeling capabilities.
Collapse
Affiliation(s)
- Gabriela Zavala
- Centro de Investigación e Innovación Biomédica (CIIB), Universidad de los Andes, Chile.
- Cells for Cells and REGENERO, The Chilean Consortium for Regenerative Medicine, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - Sergio M Viafara-García
- Centro de Investigación e Innovación Biomédica (CIIB), Universidad de los Andes, Chile.
- Cells for Cells and REGENERO, The Chilean Consortium for Regenerative Medicine, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - Javier Novoa
- Centro de Investigación e Innovación Biomédica (CIIB), Universidad de los Andes, Chile.
- Cells for Cells and REGENERO, The Chilean Consortium for Regenerative Medicine, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - Carmen Hidalgo
- Centro de Investigación e Innovación Biomédica (CIIB), Universidad de los Andes, Chile.
- Cells for Cells and REGENERO, The Chilean Consortium for Regenerative Medicine, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - Ingrid Contardo
- Centro de Investigación e Innovación Biomédica (CIIB), Universidad de los Andes, Chile.
- Facultad de Medicina, Escuela de Nutrición y Dietética, Biopolymer Research & Engineering Laboratory (BiopREL), Universidad de los Andes, Chile
| | - Paulo Díaz-Calderón
- Centro de Investigación e Innovación Biomédica (CIIB), Universidad de los Andes, Chile.
- Facultad de Medicina, Escuela de Nutrición y Dietética, Biopolymer Research & Engineering Laboratory (BiopREL), Universidad de los Andes, Chile
| | | | - Maroun Khoury
- Centro de Investigación e Innovación Biomédica (CIIB), Universidad de los Andes, Chile.
- Cells for Cells and REGENERO, The Chilean Consortium for Regenerative Medicine, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - Juan Pablo Acevedo
- Centro de Investigación e Innovación Biomédica (CIIB), Universidad de los Andes, Chile.
- Cells for Cells and REGENERO, The Chilean Consortium for Regenerative Medicine, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| |
Collapse
|
35
|
Valladares N, Cabrero Montes MA, Jacobo-Jimenez GJ, Zavala-Cerna MG. Rapid Recovery after Reparation of Full-Thickness Chondral Defects of the Knee with the Use of Hyaluronan (HA)-Based 3-D Scaffold. J Funct Biomater 2023; 14:491. [PMID: 37888156 PMCID: PMC10607491 DOI: 10.3390/jfb14100491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 09/19/2023] [Accepted: 09/21/2023] [Indexed: 10/28/2023] Open
Abstract
Articular cartilage injuries are found in up to 60% of patients who undergo an arthroscopic knee procedure, and those that totally affect articular cartilage (grade IV) have limited regenerative capacity and extended time for recovery. 3-D scaffolds represent a novel solution to address this type of injury. Our purpose was to analyze the MRI findings and functional status of patients that underwent repair of chondral defects either by microfractures or Hyaluronan (HA) 3-D scaffolding. We conducted a retrospective study of patients with chondral defects. The outcomes analyzed in this study included anatomical changes evaluated by the Henderson score (based on MRI findings) at baseline, 6, and 12 months after surgery, and improvement in functionality evaluated by the Modified Cincinnati Knee Rating System (MCKRS) at baseline and 6 months after surgery. Clinical and demographic characteristics were similar for both groups. There was a statistically significant improvement in Henderson score for the 3-D scaffold-treated group at 6 months versus the microfracture group (p < 0.0001). Improvement in functionality, measured by the MCKRS, was more frequently found in the 3-D scaffold-treated group. In conclusion, the use of HA 3-D scaffolding was superior, with faster recovery evident 6 months after the surgery that progressed to full recovery in all patients a year after surgery. Future studies with a randomized design might help to support our findings. This study provides level III evidence.
Collapse
Affiliation(s)
| | | | | | - Maria G Zavala-Cerna
- Laboratorio de Investigación en Inmunología, Unidad Académica Ciencias de la Salud, Universidad Autónoma de Guadalajara, Zapopan 45129, Mexico
| |
Collapse
|
36
|
Lázár I, Čelko L, Menelaou M. Aerogel-Based Materials in Bone and Cartilage Tissue Engineering-A Review with Future Implications. Gels 2023; 9:746. [PMID: 37754427 PMCID: PMC10530393 DOI: 10.3390/gels9090746] [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: 08/09/2023] [Revised: 09/09/2023] [Accepted: 09/11/2023] [Indexed: 09/28/2023] Open
Abstract
Aerogels are fascinating solid materials known for their highly porous nanostructure and exceptional physical, chemical, and mechanical properties. They show great promise in various technological and biomedical applications, including tissue engineering, and bone and cartilage substitution. To evaluate the bioactivity of bone substitutes, researchers typically conduct in vitro tests using simulated body fluids and specific cell lines, while in vivo testing involves the study of materials in different animal species. In this context, our primary focus is to investigate the applications of different types of aerogels, considering their specific materials, microstructure, and porosity in the field of bone and cartilage tissue engineering. From clinically approved materials to experimental aerogels, we present a comprehensive list and summary of various aerogel building blocks and their biological activities. Additionally, we explore how the complexity of aerogel scaffolds influences their in vivo performance, ranging from simple single-component or hybrid aerogels to more intricate and organized structures. We also discuss commonly used formulation and drying methods in aerogel chemistry, including molding, freeze casting, supercritical foaming, freeze drying, subcritical, and supercritical drying techniques. These techniques play a crucial role in shaping aerogels for specific applications. Alongside the progress made, we acknowledge the challenges ahead and assess the near and far future of aerogel-based hard tissue engineering materials, as well as their potential connection with emerging healing techniques.
Collapse
Affiliation(s)
- István Lázár
- Department of Inorganic and Analytical Chemistry, University of Debrecen, Egyetem tér 1, 4032 Debrecen, Hungary
| | - Ladislav Čelko
- Central European Institute of Technology, Brno University of Technology, Purkynova 656/123, 612 00 Brno, Czech Republic;
| | - Melita Menelaou
- Department of Chemical Engineering, Cyprus University of Technology, 30 Arch. Kyprianos Str., Limassol 3036, Cyprus
| |
Collapse
|
37
|
Zhang R, Chang SJ, Jing Y, Wang L, Chen CJ, Liu JT. Application of chitosan with different molecular weights in cartilage tissue engineering. Carbohydr Polym 2023; 314:120890. [PMID: 37173038 DOI: 10.1016/j.carbpol.2023.120890] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2023] [Revised: 03/27/2023] [Accepted: 04/04/2023] [Indexed: 05/15/2023]
Abstract
Cartilage tissue engineering involves the invention of novel implantable cartilage replacement materials to help heal cartilage injuries that do not heal themselves, aiming to overcome the shortcomings of current clinical cartilage treatments. Chitosan has been widely used in cartilage tissue engineering because of its similar structure to glycine aminoglycan, which is widely distributed in connective tissues. The molecular weight, as an important structural parameter of chitosan, affects not only the method of chitosan composite scaffold preparation but also the effect on cartilage tissue healing. Thus, this review identifies methods for the preparation of chitosan composite scaffolds with low, medium and high molecular weights, as well as a range of chitosan molecular weights appropriate for cartilage tissue repair, by summarizing the application of different molecular weights of chitosan in cartilage repair in recent years.
Collapse
Affiliation(s)
- Runjie Zhang
- Research Center for Materials Science and Opti-Electronic Technology, College of Materials Science and Opti-Electronic Technology, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shwu Jen Chang
- Department of Biomedical Engineering, I-Shou University, Kaohsiung City 82445, Taiwan
| | - Yanzhen Jing
- Research Center for Materials Science and Opti-Electronic Technology, College of Materials Science and Opti-Electronic Technology, University of Chinese Academy of Sciences, Beijing 100049, China
| | - LiYuan Wang
- Research Center for Materials Science and Opti-Electronic Technology, College of Materials Science and Opti-Electronic Technology, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ching-Jung Chen
- Research Center for Materials Science and Opti-Electronic Technology, School of Opto-Electronic Technology, University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Jen-Tsai Liu
- Research Center for Materials Science and Opti-Electronic Technology, College of Materials Science and Opti-Electronic Technology, University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
38
|
Ghandforoushan P, Alehosseini M, Golafshan N, Castilho M, Dolatshahi-Pirouz A, Hanaee J, Davaran S, Orive G. Injectable hydrogels for cartilage and bone tissue regeneration: A review. Int J Biol Macromol 2023; 246:125674. [PMID: 37406921 DOI: 10.1016/j.ijbiomac.2023.125674] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 06/29/2023] [Accepted: 07/01/2023] [Indexed: 07/07/2023]
Abstract
Annually, millions of patients suffer from irreversible injury owing to the loss or failure of an organ or tissue caused by accident, aging, or disease. The combination of injectable hydrogels and the science of stem cells have emerged to address this persistent issue in society by generating minimally invasive treatments to augment tissue function. Hydrogels are composed of a cross-linked network of polymers that exhibit a high-water retention capacity, thereby mimicking the wet environment of native cells. Due to their inherent mechanical softness, hydrogels can be used as needle-injectable stem cell carrier materials to mend tissue defects. Hydrogels are made of different natural or synthetic polymers, displaying a broad portfolio of eligible properties, which include biocompatibility, low cytotoxicity, shear-thinning properties as well as tunable biological and physicochemical properties. Presently, novel ongoing developments and native-like hydrogels are increasingly being used broadly to improve the quality of life of those with disabling tissue-related diseases. The present review outlines various future and in-vitro applications of injectable hydrogel-based biomaterials, focusing on the newest ongoing developments of in-situ forming injectable hydrogels for bone and cartilage tissue engineering purposes.
Collapse
Affiliation(s)
- Parisa Ghandforoushan
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tabriz University of Medical Science, Tabriz, Iran; Clinical Research Development, Unit of Tabriz Valiasr Hospital, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Morteza Alehosseini
- Department of Health Technology, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark
| | - Nasim Golafshan
- Department of Orthopedics, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Miguel Castilho
- Department of Orthopedics, University Medical Center Utrecht, Utrecht, the Netherlands; Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands
| | | | - Jalal Hanaee
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tabriz University of Medical Science, Tabriz, Iran
| | - Soodabeh Davaran
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tabriz University of Medical Science, Tabriz, Iran
| | - Gorka Orive
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country UPV/EHU Paseo de la Universidad 7, 01006 Vitoria-Gasteiz, Spain; Networking Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Vitoria-Gasteiz, Spain; Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain; University Institute for Regenerative Medicine and Oral Implantology - UIRMI (UPV/EHU-Fundación Eduardo Anitua), Vitoria, Spain; University of the Basque Country, Spain.
| |
Collapse
|
39
|
Patnaik R, Jannati S, Sivani BM, Rizzo M, Naidoo N, Banerjee Y. Efficient Generation of Chondrocytes From Bone Marrow-Derived Mesenchymal Stem Cells in a 3D Culture System: Protocol for a Practical Model for Assessing Anti-Inflammatory Therapies. JMIR Res Protoc 2023; 12:e42964. [PMID: 37505889 PMCID: PMC10437129 DOI: 10.2196/42964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 04/17/2023] [Accepted: 05/05/2023] [Indexed: 07/29/2023] Open
Abstract
BACKGROUND Chondrocytes are the primary cells responsible for maintaining cartilage integrity and function. Their role in cartilage homeostasis and response to inflammation is crucial for understanding the progression and potential therapeutic interventions for various cartilage-related disorders. Developing an accessible and cost-effective model to generate viable chondrocytes and to assess their response to different bioactive compounds can significantly advance our knowledge of cartilage biology and contribute to the discovery of novel therapeutic approaches. OBJECTIVE We developed a novel, streamlined protocol for generating chondrocytes from bone marrow-derived mesenchymal stem cells (BMSCs) in a 3D culture system that offers significant implications for the study of cartilage biology and the discovery of potential therapeutic interventions for cartilage-related and associated disorders. METHODS We developed a streamlined protocol for generating chondrocytes from BMSCs in a 3D culture system using an "in-tube" culture approach. This simple pellet-based 3D culture system allows for cell aggregation and spheroid formation, facilitating cell-cell and cell-extracellular matrix interactions that better mimic the in vivo cellular environment compared with 2D monolayer cultures. A proinflammatory chondrocyte model was created by treating the chondrocytes with lipopolysaccharide and was subsequently used to evaluate the anti-inflammatory effects of vitamin D, curcumin, and resveratrol. RESULTS The established protocol successfully generated a large quantity of viable chondrocytes, characterized by alcian blue and toluidine blue staining, and demonstrated versatility in assessing the anti-inflammatory effects of various bioactive compounds. The chondrocytes exhibited reduced inflammation, as evidenced by the decreased tumor necrosis factor-α levels, in response to vitamin D, curcumin, and resveratrol treatment. CONCLUSIONS Our novel protocol offers an accessible and cost-effective approach for generating chondrocytes from BMSCs and for evaluating potential therapeutic leads in the context of inflammatory chondrocyte-related diseases. Although our approach has several advantages, further investigation is required to address its limitations, such as the potential differences between chondrocytes generated using our protocol and those derived from other established methods, and to refine the model for broader applicability and clinical translation.
Collapse
Affiliation(s)
- Rajashree Patnaik
- Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, United Arab Emirates
| | - Shirin Jannati
- Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, United Arab Emirates
| | - Bala Mohan Sivani
- Department of Molecular Biology, Lund University, Lund, Lund, Sweden
| | - Manfredi Rizzo
- Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, United Arab Emirates
| | - Nerissa Naidoo
- Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, United Arab Emirates
| | - Yajnavalka Banerjee
- Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, United Arab Emirates
| |
Collapse
|
40
|
Cong B, Sun T, Zhao Y, Chen M. Current and Novel Therapeutics for Articular Cartilage Repair and Regeneration. Ther Clin Risk Manag 2023; 19:485-502. [PMID: 37360195 PMCID: PMC10290456 DOI: 10.2147/tcrm.s410277] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 05/28/2023] [Indexed: 06/28/2023] Open
Abstract
Articular cartilage repair is a sophisticated process that has is being recently investigated. There are several different approaches that are currently reported to promote cartilage repair, like cell-based therapies, biologics, and physical therapy. Cell-based therapies involve the using stem cells or chondrocytes, which make up cartilage, to promote the growth of new cartilage. Biologics, like growth factors, are also being applied to enhance cartilage repair. Physical therapy, like exercise and weight-bearing activities, can also be used to promote cartilage repair by inducing new cartilage growth and improving joint function. Additionally, surgical options like osteochondral autograft, autologous chondrocyte implantation, microfracture, and others are also reported for cartilage regeneration. In the current literature review, we aim to provide an up-to-date discussion about these approaches and discuss the current research status.
Collapse
Affiliation(s)
- Bo Cong
- Department of Orthopedics, Yantaishan Hospital Affiliated to Binzhou Medical University, Yantai, 264003, People’s Republic of China
- Yantai Key Laboratory for Repair and Reconstruction of Bone & Joint, Yantai, 264003, People’s Republic of China
| | - Tao Sun
- Department of Orthopedics, Yantaishan Hospital Affiliated to Binzhou Medical University, Yantai, 264003, People’s Republic of China
- Yantai Key Laboratory for Repair and Reconstruction of Bone & Joint, Yantai, 264003, People’s Republic of China
| | - Yuchi Zhao
- Department of Orthopedics, Yantaishan Hospital Affiliated to Binzhou Medical University, Yantai, 264003, People’s Republic of China
- Yantai Key Laboratory for Repair and Reconstruction of Bone & Joint, Yantai, 264003, People’s Republic of China
| | - Mingqi Chen
- Department of Orthopedics, Yantaishan Hospital Affiliated to Binzhou Medical University, Yantai, 264003, People’s Republic of China
| |
Collapse
|
41
|
Płończak M, Wasyłeczko M, Jakutowicz T, Chwojnowski A, Czubak J. Intraarticular Implantation of Autologous Chondrocytes Placed on Collagen or Polyethersulfone Scaffolds: An Experimental Study in Rabbits. Polymers (Basel) 2023; 15:polym15102360. [PMID: 37242936 DOI: 10.3390/polym15102360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 05/15/2023] [Accepted: 05/16/2023] [Indexed: 05/28/2023] Open
Abstract
Hyaline cartilage has very limited repair capability and cannot be rebuilt predictably using conventional treatments. This study presents Autologous Chondrocyte Implantation (ACI) on two different scaffolds for the treatment of lesions in hyaline cartilage in rabbits. The first one is a commercially available scaffold (Chondro-Gide) made of collagen type I/III and the second one is a polyethersulfone (PES) synthetic membrane, manufactured by phase inversion. The revolutionary idea in the present study is the fact that we used PES membranes, which have unique features and benefits that are desirable for the 3D cultivation of chondrocytes. Sixty-four White New Zealand rabbits were used in this research. Defects penetrating into the subchondral bone were filled with or without the placement of chondrocytes on collagen or PES membranes after two weeks of culture. The expression of the gene encoding type II procollagen, a molecular marker of chondrocytes, was evaluated. Elemental analysis was performed to estimate the weight of tissue grown on the PES membrane. The reparative tissue was analyzed macroscopically and histologically after surgery at 12, 25, and 52 weeks. RT-PCR analysis of the mRNA isolated from cells detached from the polysulphonic membrane revealed the expression of type II procollagen. The elementary analysis of polysulphonic membrane slices after 2 weeks of culture with chondrocytes revealed a concentration of 0.23 mg of tissue on one part of the membrane. Macroscopic and microscopic evaluation indicated that the quality of regenerated tissue was similar after the transplantation of cells placed on polysulphonic or collagen membranes. The established method for the culture and transplantation of chondrocytes placed on polysulphonic membranes resulted in the growth of the regenerated tissue, revealing the morphology of hyaline-like cartilage to be of similar quality to collagen membranes.
Collapse
Affiliation(s)
- Maciej Płończak
- Mazovia Regional Hospital John Paul II, 08-110 Siedlce, Poland
| | - Monika Wasyłeczko
- Nałęcz Institute of Biocybernetic and Biomedical Engineering, Polish Academy of Sciences, 02-109 Warsaw, Poland
| | - Tomasz Jakutowicz
- Department of Neurosurgery and Children Traumatology, Medical University of Warsaw, 02-091 Warsaw, Poland
| | - Andrzej Chwojnowski
- Nałęcz Institute of Biocybernetic and Biomedical Engineering, Polish Academy of Sciences, 02-109 Warsaw, Poland
| | - Jarosław Czubak
- Department of Orthopedics, Pediatric Orthopedics and Traumatology, Centre of Postgraduate Medical Education, Gruca Orthopaedic and Trauma Teaching Hospital, 05-402 Otwock, Poland
| |
Collapse
|
42
|
González-González AM, Cruz R, Rosales-Ibáñez R, Hernández-Sánchez F, Carrillo-Escalante HJ, Rodríguez-Martínez JJ, Velasquillo C, Talamás-Lara D, Ludert JE. In Vitro and In Vivo Evaluation of a Polycaprolactone (PCL)/Polylactic-Co-Glycolic Acid (PLGA) (80:20) Scaffold for Improved Treatment of Chondral (Cartilage) Injuries. Polymers (Basel) 2023; 15:polym15102324. [PMID: 37242899 DOI: 10.3390/polym15102324] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 05/09/2023] [Accepted: 05/10/2023] [Indexed: 05/28/2023] Open
Abstract
Articular cartilage is a specialized tissue that provides a smooth surface for joint movement and load transmission. Unfortunately, it has limited regenerative capacity. Tissue engineering, combining different cell types, scaffolds, growth factors, and physical stimulation has become an alternative for repairing and regenerating articular cartilage. Dental Follicle Mesenchymal Stem Cells (DFMSCs) are attractive candidates for cartilage tissue engineering because of their ability to differentiate into chondrocytes, on the other hand, the polymers blend like Polycaprolactone (PCL) and Poly Lactic-co-Glycolic Acid (PLGA) have shown promise given their mechanical properties and biocompatibility. In this work, the physicochemical properties of polymer blends were evaluated by Fourier Transform Infrared Spectroscopy (FTIR) and Scanning Electron Microscope (SEM) and were positive for both techniques. The DFMSCs demonstrated stemness by flow cytometry. The scaffold showed to be a non-toxic effect when we evaluated it with Alamar blue, and the samples were analyzed using SEM and phalloidin staining to evaluate cell adhesion to the scaffold. The synthesis of glycosaminoglycans was positive on the construct in vitro. Finally, the PCL/PLGA scaffold showed a better repair capacity than two commercial compounds, when tested in a chondral defect rat model. These results suggest that the PCL/PLGA (80:20) scaffold may be suitable for applications in the tissue engineering of articular hyaline cartilage.
Collapse
Affiliation(s)
- Arely M González-González
- Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies (CINVESTAV), Mexico City 07360, Mexico
- Laboratorio de Ingeniería Tisular y Medicina Traslacional, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de Mexico (UNAM), Mexico City 54090, Mexico
| | - Raymundo Cruz
- Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies (CINVESTAV), Mexico City 07360, Mexico
| | - Raúl Rosales-Ibáñez
- Laboratorio de Ingeniería Tisular y Medicina Traslacional, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de Mexico (UNAM), Mexico City 54090, Mexico
| | | | | | - Jesús Jiovanni Rodríguez-Martínez
- Laboratorio de Ingeniería Tisular y Medicina Traslacional, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de Mexico (UNAM), Mexico City 54090, Mexico
| | - Cristina Velasquillo
- Unidad de Ingeniería de Tejidos, Terapia Celular y Medicina Regenerativa, Instituto Nacional de Rehabilitación "Luis Guillermo Ibarra Ibarra", Ciudad de Mexico 14389, Mexico
| | - Daniel Talamás-Lara
- Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies (CINVESTAV), Mexico City 07360, Mexico
| | - Juan E Ludert
- Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies (CINVESTAV), Mexico City 07360, Mexico
| |
Collapse
|
43
|
Wasyłeczko M, Remiszewska E, Sikorska W, Dulnik J, Chwojnowski A. Scaffolds for Cartilage Tissue Engineering from a Blend of Polyethersulfone and Polyurethane Polymers. Molecules 2023; 28:molecules28073195. [PMID: 37049957 PMCID: PMC10095814 DOI: 10.3390/molecules28073195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 03/29/2023] [Accepted: 03/31/2023] [Indexed: 04/07/2023] Open
Abstract
In recent years, one of the main goals of cartilage tissue engineering has been to find appropriate scaffolds for hyaline cartilage regeneration, which could serve as a matrix for chondrocytes or stem cell cultures. The study presents three types of scaffolds obtained from a blend of polyethersulfone (PES) and polyurethane (PUR) by a combination of wet-phase inversion and salt-leaching methods. The nonwovens made of gelatin and sodium chloride (NaCl) were used as precursors of macropores. Thus, obtained membranes were characterized by a suitable structure. The top layers were perforated, with pores over 20 µm, which allows cells to enter the membrane. The use of a nonwoven made it possible to develop a three-dimensional network of interconnected macropores that is required for cell activity and mobility. Examination of wettability (contact angle, swelling ratio) showed a hydrophilic nature of scaffolds. The mechanical test showed that the scaffolds were suitable for knee joint applications (stress above 10 MPa). Next, the scaffolds underwent a degradation study in simulated body fluid (SBF). Weight loss after four weeks and changes in structure were assessed using scanning electron microscopy (SEM) and MeMoExplorer Software, a program that estimates the size of pores. The porosity measurements after degradation confirmed an increase in pore size, as expected. Hydrolysis was confirmed by Fourier-transform infrared spectroscopy (FT-IR) analysis, where the disappearance of ester bonds at about 1730 cm−1 wavelength is noticeable after degradation. The obtained results showed that the scaffolds meet the requirements for cartilage tissue engineering membranes and should undergo further testing on an animal model.
Collapse
Affiliation(s)
- Monika Wasyłeczko
- Nalecz Institute of Biocybernetics and Biomedical Engineering, Polish Academy of Sciences, Trojdena 4, 02-109 Warsaw, Poland
| | - Elżbieta Remiszewska
- Nalecz Institute of Biocybernetics and Biomedical Engineering, Polish Academy of Sciences, Trojdena 4, 02-109 Warsaw, Poland
| | - Wioleta Sikorska
- Nalecz Institute of Biocybernetics and Biomedical Engineering, Polish Academy of Sciences, Trojdena 4, 02-109 Warsaw, Poland
| | - Judyta Dulnik
- Institute of Fundamental Technological Research Polish Academy of Sciences, Laboratory of Polymers and Biomaterials, Pawińskiego 5b, 02-106 Warsaw, Poland
| | - Andrzej Chwojnowski
- Nalecz Institute of Biocybernetics and Biomedical Engineering, Polish Academy of Sciences, Trojdena 4, 02-109 Warsaw, Poland
| |
Collapse
|
44
|
Gao LL, Wei Y, Tan YS, Li RX, Zhang CQ, Gao H. Irrigating degradation properties of silk fibroin-collagen type II composite cartilage scaffold in vitro and in vivo. BIOMATERIALS ADVANCES 2023; 149:213389. [PMID: 36965402 DOI: 10.1016/j.bioadv.2023.213389] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Revised: 03/07/2023] [Accepted: 03/13/2023] [Indexed: 03/17/2023]
Abstract
Silk fibroin-collagen type II scaffolds are promising in cartilage tissue engineering due to their suitable biological functionality to promote proliferation of chondrocytes in vitro. However, their degradation properties, which are of crucial importance as scaffold degradation should consistent with the new tissue formation process, are still unknown. In this study, degradability of silk fibroin-collagen type II cartilage scaffolds was probed both in vitro and in vivo. In vitro degradation experiments show that the scaffolds decreased 32.25 % ± 0.62 %, 34.27 % ± 0.96 %, 36.27 % ± 2.39 % in weight after 8 weeks of degradation at the irrigation velocity of 0 mL/min, 7.89 mL/min and 15.79 mL/min. The degradation ratio, which increases with time and increasing irrigation velocity, is described by combining the built mathematic model and finite element modeling method. The scaffolds after 8 weeks of degradation in vitro keep their mechanical structural integrity to support new tissues. In vivo degradation experiments conducted in rabbits further show that the scaffolds degrade gradually, be absorbed with time and finally collapse in structure. The degradation process is accompanied by the growth of fibrous tissues and the scaffold is filled by fibrous tissues after 12 weeks of implantation. Immunohistology analysis shows that the inflammation caused by scaffolds is controllable and gradually alleviates with time. To sum up, silk fibroin-collagen type II cartilage scaffolds, which show suitable mechanical properties and biocompatibility during degradation in vitro and in vivo, have great potential in cartilage repair. The novelty of the study is that it not only introduces a mathematical model to predict the irrigation degradation ratio, but also provides experimental degradation data support for clinical application of silk fibroin-collagen type II cartilage scaffolds.
Collapse
Affiliation(s)
- Li-Lan Gao
- Tianjin Key Laboratory for Advanced Mechatronic System Design and Intelligent Control, School of Mechanical Engineering, Tianjin University of Technology, Tianjin, China; National Demonstration Center for Experimental Mechanical and Electrical Engineering Education, Tianjin University of Technology, Tianjin, China
| | - Ying Wei
- Tianjin Key Laboratory for Advanced Mechatronic System Design and Intelligent Control, School of Mechanical Engineering, Tianjin University of Technology, Tianjin, China; National Demonstration Center for Experimental Mechanical and Electrical Engineering Education, Tianjin University of Technology, Tianjin, China
| | - Yan-Song Tan
- Tianjin Key Laboratory for Advanced Mechatronic System Design and Intelligent Control, School of Mechanical Engineering, Tianjin University of Technology, Tianjin, China; National Demonstration Center for Experimental Mechanical and Electrical Engineering Education, Tianjin University of Technology, Tianjin, China.
| | - Rui-Xin Li
- Tianjin Stomatological Hospital, Tianjin, China.
| | - Chun-Qiu Zhang
- Tianjin Key Laboratory for Advanced Mechatronic System Design and Intelligent Control, School of Mechanical Engineering, Tianjin University of Technology, Tianjin, China; National Demonstration Center for Experimental Mechanical and Electrical Engineering Education, Tianjin University of Technology, Tianjin, China.
| | - Hong Gao
- School of Chemical Engineering and Technology, Tianjin University, Tianjin, China
| |
Collapse
|
45
|
Horvath-Pereira BDO, Almeida GHDR, da Silva Júnior LN, do Nascimento PG, Horvath Pereira BDO, Fireman JVBT, Pereira MLDRF, Carreira ACO, Miglino MA. Biomaterials for Testicular Bioengineering: How far have we come and where do we have to go? Front Endocrinol (Lausanne) 2023; 14:1085872. [PMID: 37008920 PMCID: PMC10060902 DOI: 10.3389/fendo.2023.1085872] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 02/24/2023] [Indexed: 03/18/2023] Open
Abstract
Traditional therapeutic interventions aim to restore male fertile potential or preserve sperm viability in severe cases, such as semen cryopreservation, testicular tissue, germ cell transplantation and testicular graft. However, these techniques demonstrate several methodological, clinical, and biological limitations, that impact in their results. In this scenario, reproductive medicine has sought biotechnological alternatives applied for infertility treatment, or to improve gamete preservation and thus increase reproductive rates in vitro and in vivo. One of the main approaches employed is the biomimetic testicular tissue reconstruction, which uses tissue-engineering principles and methodologies. This strategy pursues to mimic the testicular microenvironment, simulating physiological conditions. Such approach allows male gametes maintenance in culture or produce viable grafts that can be transplanted and restore reproductive functions. In this context, the application of several biomaterials have been proposed to be used in artificial biological systems. From synthetic polymers to decellularized matrixes, each biomaterial has advantages and disadvantages regarding its application in cell culture and tissue reconstruction. Therefore, the present review aims to list the progress that has been made and the continued challenges facing testicular regenerative medicine and the preservation of male reproductive capacity, based on the development of tissue bioengineering approaches for testicular tissue microenvironment reconstruction.
Collapse
Affiliation(s)
| | | | | | - Pedro Gabriel do Nascimento
- Department of Surgery, School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, Brazil
| | | | | | | | - Ana Claudia Oliveira Carreira
- Department of Surgery, School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, Brazil
- Centre for Natural and Human Sciences, Federal University of ABC, São Paulo, Brazil
| | - Maria Angelica Miglino
- Department of Surgery, School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
46
|
Samie M, Khan AF, Rahman SU, Iqbal H, Yameen MA, Chaudhry AA, Galeb HA, Halcovitch NR, Hardy JG. Drug/bioactive eluting chitosan composite foams for osteochondral tissue engineering. Int J Biol Macromol 2023; 229:561-574. [PMID: 36587649 DOI: 10.1016/j.ijbiomac.2022.12.293] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Revised: 12/19/2022] [Accepted: 12/25/2022] [Indexed: 12/30/2022]
Abstract
Joint defects associated with a variety of etiologies often extend deep into the subchondral bone leading to functional impairment and joint immobility, and it is a very challenging task to regenerate the bone-cartilage interface offering significant opportunities for biomaterial-based interventions to improve the quality of life of patients. Herein drug-/bioactive-loaded porous tissue scaffolds incorporating nano-hydroxyapatite (nHAp), chitosan (CS) and either hydroxypropyl methylcellulose (HPMC) or Bombyx mori silk fibroin (SF) are fabricated through freeze drying method as subchondral bone substitute. A combination of spectroscopy and microscopy (Fourier transform infrared (FTIR) spectroscopy, scanning electron microscopy (SEM), X-ray diffraction (XRD), energy dispersive X-ray (EDX), and X-ray fluorescence (XRF) were used to analyze the structure of the porous biomaterials. The compressive mechanical properties of these scaffolds are biomimetic of cancellous bone tissues and capable of releasing drugs/bioactives (exemplified with triamcinolone acetonide, TA, or transforming growth factor-β1, TGF-β1, respectively) over a period of days. Mouse preosteoblast MC3T3-E1 cells were observed to adhere and proliferate on the tissue scaffolds as confirmed by the cell attachment, live-dead assay and alamarBlue™ assay. Interestingly, RT-qPCR analysis showed that the TA downregulated inflammatory biomarkers and upregulated the bone-specific biomarkers, suggesting such tissue scaffolds have long-term potential for clinical application.
Collapse
Affiliation(s)
- Muhammad Samie
- Interdisciplinary Research Centre in Biomedical Materials, COMSATS University Islamabad, Lahore Campus, 54000, Pakistan; Department of Pharmacy, COMSATS University Islamabad, Abbottabad Campus, 22060, Pakistan; Department of Chemistry, Lancaster University, Lancaster, Lancashire LA1 4YB, United Kingdom; Materials Science Institute, Lancaster University, Lancaster, Lancashire LA1 4YW, United Kingdom; Institute of Pharmaceutical Sciences, Khyber Medical University, Peshawar, Khyber Pakhtunkhwa 25100, Pakistan.
| | - Ather Farooq Khan
- Interdisciplinary Research Centre in Biomedical Materials, COMSATS University Islamabad, Lahore Campus, 54000, Pakistan
| | - Saeed Ur Rahman
- Institute of Basic Medical Sciences, Khyber Medical University, Peshawar, Khyber Pakhtunkhwa 25100, Pakistan
| | - Haffsah Iqbal
- Interdisciplinary Research Centre in Biomedical Materials, COMSATS University Islamabad, Lahore Campus, 54000, Pakistan
| | - Muhammad Arfat Yameen
- Department of Pharmacy, COMSATS University Islamabad, Abbottabad Campus, 22060, Pakistan
| | - Aqif Anwar Chaudhry
- Interdisciplinary Research Centre in Biomedical Materials, COMSATS University Islamabad, Lahore Campus, 54000, Pakistan
| | - Hanaa A Galeb
- Department of Chemistry, Lancaster University, Lancaster, Lancashire LA1 4YB, United Kingdom; Department of Chemistry, Science and Arts College, Rabigh Campus, King Abdulaziz University, 21577 Jeddah, Saudi Arabia
| | - Nathan R Halcovitch
- Department of Chemistry, Lancaster University, Lancaster, Lancashire LA1 4YB, United Kingdom
| | - John G Hardy
- Department of Chemistry, Lancaster University, Lancaster, Lancashire LA1 4YB, United Kingdom; Materials Science Institute, Lancaster University, Lancaster, Lancashire LA1 4YW, United Kingdom.
| |
Collapse
|
47
|
Sun W, Gregory DA, Zhao X. Designed peptide amphiphiles as scaffolds for tissue engineering. Adv Colloid Interface Sci 2023; 314:102866. [PMID: 36898186 DOI: 10.1016/j.cis.2023.102866] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 02/22/2023] [Accepted: 02/24/2023] [Indexed: 03/03/2023]
Abstract
Peptide amphiphiles (PAs) are peptide-based molecules that contain a peptide sequence as a head group covalently conjugated to a hydrophobic segment, such as lipid tails. They can self-assemble into well-ordered supramolecular nanostructures such as micelles, vesicles, twisted ribbons and nanofibers. In addition, the diversity of natural amino acids gives the possibility to produce PAs with different sequences. These properties along with their biocompatibility, biodegradability and a high resemblance to native extracellular matrix (ECM) have resulted in PAs being considered as ideal scaffold materials for tissue engineering (TE) applications. This review introduces the 20 natural canonical amino acids as building blocks followed by highlighting the three categories of PAs: amphiphilic peptides, lipidated peptide amphiphiles and supramolecular peptide amphiphile conjugates, as well as their design rules that dictate the peptide self-assembly process. Furthermore, 3D bio-fabrication strategies of PAs hydrogels are discussed and the recent advances of PA-based scaffolds in TE with the emphasis on bone, cartilage and neural tissue regeneration both in vitro and in vivo are considered. Finally, future prospects and challenges are discussed.
Collapse
Affiliation(s)
- Weizhen Sun
- School of Pharmacy, Changzhou University, Changzhou 213164, China; Department of Chemical and Biological Engineering, University of Sheffield, Sheffield S1 3JD, UK
| | - David Alexander Gregory
- Department of Chemical and Biological Engineering, University of Sheffield, Sheffield S1 3JD, UK; Department of Material Science and Engineering, University of Sheffield, Sheffield S3 7HQ, UK
| | - Xiubo Zhao
- School of Pharmacy, Changzhou University, Changzhou 213164, China; Department of Chemical and Biological Engineering, University of Sheffield, Sheffield S1 3JD, UK.
| |
Collapse
|
48
|
Taheri S, Ghazali ZS, Montazeri L, Ebrahim FA, Javadpour J, Kamguyan K, Thormann E, Renaud P, Bonakdar S. Engineered substrates incapable of induction of chondrogenic differentiation compared to the chondrocyte imprinted substrates. Biomed Mater 2023; 18. [PMID: 36693281 DOI: 10.1088/1748-605x/acb5d7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 01/24/2023] [Indexed: 01/26/2023]
Abstract
It is well established that surface topography can affect cell functions. However, finding a reproducible and reliable method for regulating stem cell behavior is still under investigation. It has been shown that cell imprinted substrates contain micro- and nanoscale structures of the cell membrane that serve as hierarchical substrates, can successfully alter stem cell fate. This study investigated the effect of the overall cell shape by fabricating silicon wafers containing pit structure in the average size of spherical-like chondrocytes using photolithography technique. We also used chondrocyte cell line (C28/I2) with spindle-like shape to produce cell imprinted substrates. The effect of all substrates on the differentiation of adipose-derived mesenchymal stem cells (ADSCs) has been studied. The AFM and scanning electron microscopy images of the prepared substrates demonstrated that the desired shapes were successfully transferred to the substrates. Differentiation of ADSCs was investigated by immunostaining for mature chondrocyte marker, collagen II, and gene expression of collagen II, Sox9, and aggrecan markers. C28/I2 imprinted substrate could effectively enhanced chondrogenic differentiation compared to regular pit patterns on the wafer. It can be concluded that cell imprinted substrates can induce differentiation signals better than engineered lithographic substrates. The nanostructures on the cell-imprinted patterns play a crucial role in harnessing cell fate. Therefore, the patterns must include the nano-topographies to have reliable and reproducible engineered substrates.
Collapse
Affiliation(s)
- Shiva Taheri
- National Cell Bank Department, Iran Pasteur Institute, Tehran, Iran.,School of Metallurgy and Materials Engineering, Iran University of Science and Technology, Tehran, Iran
| | - Zahra Sadat Ghazali
- Department of Biomedical Engineering, Amirkabir University of Technology, Tehran, Iran
| | - Leila Montazeri
- Department of Cell Engineering, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | | | - Jafar Javadpour
- School of Metallurgy and Materials Engineering, Iran University of Science and Technology, Tehran, Iran
| | - Khorshid Kamguyan
- Department of Health Technology, Technical University of Denmark, 2800 Kongens Lyngby, Denmark
| | - Esben Thormann
- Department of Chemistry, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark
| | - Philippe Renaud
- STI-IMT-LMIS4, École Polytechnique Fédérale de Lausanne, Station 17, 1015 Lausanne, Switzerland
| | - Shahin Bonakdar
- National Cell Bank Department, Iran Pasteur Institute, Tehran, Iran
| |
Collapse
|
49
|
Cordeiro R, Alvites RD, Sousa AC, Lopes B, Sousa P, Maurício AC, Alves N, Moura C. Cellulose-Based Scaffolds: A Comparative Study for Potential Application in Articular Cartilage. Polymers (Basel) 2023; 15:781. [PMID: 36772083 PMCID: PMC9919712 DOI: 10.3390/polym15030781] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 02/01/2023] [Accepted: 02/01/2023] [Indexed: 02/09/2023] Open
Abstract
Osteoarthritis is a highly prevalent disease worldwide that leads to cartilage loss. Tissue engineering, involving scaffolds, cells, and stimuli, has shown to be a promising strategy for its repair. Thus, this study aims to manufacture and characterise different scaffolds with poly(ε-caprolactone) (PCL) with commercial cellulose (microcrystalline (McC) and methyl cellulose (MC) or cellulose from agro-industrial residues (corncob (CcC)) and at different percentages, 1%, 2%, and 3%. PCL scaffolds were used as a control. Morphologically, the produced scaffolds presented porosities within the desired for cell incorporation (57% to 65%). When submitted to mechanical tests, the incorporation of cellulose affects the compression resistance of the majority of scaffolds. Regarding tensile strength, McC2% showed the highest values. It was proven that all manufactured scaffolds suffered degradation after 7 days of testing because of enzymatic reactions. This degradation may be due to the dissolution of PCL in the organic solvent. Biological tests revealed that PCL, CcC1%, and McC3% are the best materials to combine with human dental pulp stem/stromal cells. Overall, results suggest that cellulose incorporation in PCL scaffolds promotes cellular adhesion/proliferation. Methyl cellulose scaffolds demonstrated some advantageous compressive properties (closer to native cartilaginous tissue) to proceed to further studies for application in cartilage repair.
Collapse
Affiliation(s)
- Rachel Cordeiro
- Centre for Rapid and Sustainable Product Development, Polytechnic of Leiria, 2430-028 Marinha Grande, Portugal
- Veterinary Clinics Department, Abel Salazar Biomedical Sciences Institute (ICBAS), University of Porto (UP), Rua de Jorge Viterbo Ferreira, No. 228, 4050-313 Porto, Portugal
| | - Rui D. Alvites
- Veterinary Clinics Department, Abel Salazar Biomedical Sciences Institute (ICBAS), University of Porto (UP), Rua de Jorge Viterbo Ferreira, No. 228, 4050-313 Porto, Portugal
- Animal Science Studies Centre (CECA), Agroenvironment, Technologies and Sciences Institute (ICETA), University of Porto, Rua D. Manuel II, Apartado 55142, 4051-401 Porto, Portugal
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), 1300-477 Lisbon, Portugal
| | - Ana C. Sousa
- Veterinary Clinics Department, Abel Salazar Biomedical Sciences Institute (ICBAS), University of Porto (UP), Rua de Jorge Viterbo Ferreira, No. 228, 4050-313 Porto, Portugal
- Animal Science Studies Centre (CECA), Agroenvironment, Technologies and Sciences Institute (ICETA), University of Porto, Rua D. Manuel II, Apartado 55142, 4051-401 Porto, Portugal
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), 1300-477 Lisbon, Portugal
| | - Bruna Lopes
- Veterinary Clinics Department, Abel Salazar Biomedical Sciences Institute (ICBAS), University of Porto (UP), Rua de Jorge Viterbo Ferreira, No. 228, 4050-313 Porto, Portugal
- Animal Science Studies Centre (CECA), Agroenvironment, Technologies and Sciences Institute (ICETA), University of Porto, Rua D. Manuel II, Apartado 55142, 4051-401 Porto, Portugal
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), 1300-477 Lisbon, Portugal
| | - Patrícia Sousa
- Veterinary Clinics Department, Abel Salazar Biomedical Sciences Institute (ICBAS), University of Porto (UP), Rua de Jorge Viterbo Ferreira, No. 228, 4050-313 Porto, Portugal
- Animal Science Studies Centre (CECA), Agroenvironment, Technologies and Sciences Institute (ICETA), University of Porto, Rua D. Manuel II, Apartado 55142, 4051-401 Porto, Portugal
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), 1300-477 Lisbon, Portugal
| | - Ana C. Maurício
- Veterinary Clinics Department, Abel Salazar Biomedical Sciences Institute (ICBAS), University of Porto (UP), Rua de Jorge Viterbo Ferreira, No. 228, 4050-313 Porto, Portugal
- Animal Science Studies Centre (CECA), Agroenvironment, Technologies and Sciences Institute (ICETA), University of Porto, Rua D. Manuel II, Apartado 55142, 4051-401 Porto, Portugal
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), 1300-477 Lisbon, Portugal
| | - Nuno Alves
- Centre for Rapid and Sustainable Product Development, Polytechnic of Leiria, 2430-028 Marinha Grande, Portugal
- Associate Laboratory for Advanced Production and Intelligent Systems (ARISE), 4050-313 Porto, Portugal
| | - Carla Moura
- Centre for Rapid and Sustainable Product Development, Polytechnic of Leiria, 2430-028 Marinha Grande, Portugal
- Associate Laboratory for Advanced Production and Intelligent Systems (ARISE), 4050-313 Porto, Portugal
- Applied Research Institute (i2A), Polytechnic Institute of Coimbra, Rua da Misericórdia, Lagar dos Cortiços–S. Martinho do Bispo, 3045-093 Coimbra, Portugal
| |
Collapse
|
50
|
Guo X, Ma Y, Min Y, Sun J, Shi X, Gao G, Sun L, Wang J. Progress and prospect of technical and regulatory challenges on tissue-engineered cartilage as therapeutic combination product. Bioact Mater 2023; 20:501-518. [PMID: 35846847 PMCID: PMC9253051 DOI: 10.1016/j.bioactmat.2022.06.015] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 06/19/2022] [Accepted: 06/19/2022] [Indexed: 12/18/2022] Open
Abstract
Hyaline cartilage plays a critical role in maintaining joint function and pain. However, the lack of blood supply, nerves, and lymphatic vessels greatly limited the self-repair and regeneration of damaged cartilage, giving rise to various tricky issues in medicine. In the past 30 years, numerous treatment techniques and commercial products have been developed and practiced in the clinic for promoting defected cartilage repair and regeneration. Here, the current therapies and their relevant advantages and disadvantages will be summarized, particularly the tissue engineering strategies. Furthermore, the fabrication of tissue-engineered cartilage under research or in the clinic was discussed based on the traid of tissue engineering, that is the materials, seed cells, and bioactive factors. Finally, the commercialized cartilage repair products were listed and the regulatory issues and challenges of tissue-engineered cartilage repair products and clinical application would be reviewed. Tissue engineered cartilage, a promising strategy for articular cartilage repair. Nearly 20 engineered cartilage repair products in clinic based on clinical techniques. Combination product, the classification of tissue-engineered cartilage. Key regulatory compliance issues for combination products.
Collapse
Affiliation(s)
- Xiaolei Guo
- Center for Medical Device Evaluation, National Medical Products Administration, Beijing, PR China
- Corresponding author.
| | - Yuan Ma
- State Key Laboratory of Tribology, Tsinghua University, Beijing, PR China
| | - Yue Min
- Center for Medical Device Evaluation, National Medical Products Administration, Beijing, PR China
| | - Jiayi Sun
- Center for Medical Device Evaluation, National Medical Products Administration, Beijing, PR China
| | - Xinli Shi
- Center for Medical Device Evaluation, National Medical Products Administration, Beijing, PR China
- Corresponding author. Center for Medical Device Evaluation, National Medical Products Administration, Beijing, 100081, PR China
| | - Guobiao Gao
- Center for Medical Device Evaluation, National Medical Products Administration, Beijing, PR China
| | - Lei Sun
- Center for Medical Device Evaluation, National Medical Products Administration, Beijing, PR China
| | - Jiadao Wang
- State Key Laboratory of Tribology, Tsinghua University, Beijing, PR China
- Corresponding author. State Key Laboratory of Tribology, Tsinghua University, Beijing 100084, China.
| |
Collapse
|