1
|
Wu X, Zhu L, Xia L, Gui L, Cong W. The phenotypic characterization of mouse floxed Fam20b chondrocytes, a novel articular cartilage-derived cell line with differentiation potential. Regen Ther 2025; 29:108-116. [PMID: 40162017 PMCID: PMC11953959 DOI: 10.1016/j.reth.2025.02.003] [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: 12/03/2024] [Revised: 01/21/2025] [Accepted: 02/12/2025] [Indexed: 04/02/2025] Open
Abstract
FAM20B is a newly identified kinase involved in proteoglycan biosynthesis. Inactivation of Fam20b in joint cartilage leads to chondrosarcoma. Generation of chondrocytes cell line carrying floxed Fam20b allele can offer a valuable tool for the study of the role of Fam20b as well as the molecular events in chondrocyte biology and cartilage diseases. The limitations in the primary culture of chondrocytes necessitate the development of chondrocyte cell line. In this study, we established and characterized the immortalized mouse floxed Fam20b chondrocyte cell line. The primary mouse floxed Fam20b chondrocytes were isolated from the articular cartilage of knee joints and immortalized using lentivirus containing Simian Virus 40 T-antigen (SV40 T-Ag). The immortalized cell line was verified by genomic integration of SV40 T-Ag and proliferated at a high rate relative to their primary counterparts. The immortalized chondrocyte cell line not only retained the typical ultrastructural morphology and important phenotypic characteristics of articular cartilage, but also possessed strong differentiation potential upon three-dimensional pellet culture. Thus, we, for the first time, describe the development of immortalized mouse floxed Fam20b chondrocytes and present an alternative for the limited number of articular chondrocyte cell lines for the study of cartilage biology.
Collapse
Affiliation(s)
- Xiaoyuan Wu
- School of Stomatology, Dalian Medical University, Dalian, Liaoning, China
- Department of Periodontics, Affiliated Stomatological Hospital of Suzhou Vocational Health College, Suzhou, Jiangsu, China
| | - Lei Zhu
- School of Stomatology, Dalian Medical University, Dalian, Liaoning, China
| | - Lifeng Xia
- School of Stomatology, Dalian Medical University, Dalian, Liaoning, China
| | - Lin Gui
- School of Stomatology, Dalian Medical University, Dalian, Liaoning, China
| | - Wei Cong
- School of Stomatology, Dalian Medical University, Dalian, Liaoning, China
- Academican Laboratory of Immune and Oral Development & Regeneration, Dalian Medical University, Dalian, Liaoning, China
| |
Collapse
|
2
|
Grzelak A, Hnydka A, Higuchi J, Michalak A, Tarczynska M, Gaweda K, Klimek K. Recent Achievements in the Development of Biomaterials Improved with Platelet Concentrates for Soft and Hard Tissue Engineering Applications. Int J Mol Sci 2024; 25:1525. [PMID: 38338805 PMCID: PMC10855389 DOI: 10.3390/ijms25031525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 01/19/2024] [Accepted: 01/23/2024] [Indexed: 02/12/2024] Open
Abstract
Platelet concentrates such as platelet-rich plasma, platelet-rich fibrin or concentrated growth factors are cost-effective autologous preparations containing various growth factors, including platelet-derived growth factor, transforming growth factor β, insulin-like growth factor 1 and vascular endothelial growth factor. For this reason, they are often used in regenerative medicine to treat wounds, nerve damage as well as cartilage and bone defects. Unfortunately, after administration, these preparations release growth factors very quickly, which lose their activity rapidly. As a consequence, this results in the need to repeat the therapy, which is associated with additional pain and discomfort for the patient. Recent research shows that combining platelet concentrates with biomaterials overcomes this problem because growth factors are released in a more sustainable manner. Moreover, this concept fits into the latest trends in tissue engineering, which include biomaterials, bioactive factors and cells. Therefore, this review presents the latest literature reports on the properties of biomaterials enriched with platelet concentrates for applications in skin, nerve, cartilage and bone tissue engineering.
Collapse
Affiliation(s)
- Agnieszka Grzelak
- Chair and Department of Biochemistry and Biotechnology, Medical University of Lublin, Chodzki Street 1, 20-093 Lublin, Poland; (A.G.); (A.H.)
| | - Aleksandra Hnydka
- Chair and Department of Biochemistry and Biotechnology, Medical University of Lublin, Chodzki Street 1, 20-093 Lublin, Poland; (A.G.); (A.H.)
| | - Julia Higuchi
- Laboratory of Nanostructures, Institute of High Pressure Physics, Polish Academy of Sciences, Prymasa Tysiaclecia Avenue 98, 01-142 Warsaw, Poland;
| | - Agnieszka Michalak
- Independent Laboratory of Behavioral Studies, Medical University of Lublin, Chodzki 4 a Street, 20-093 Lublin, Poland;
| | - Marta Tarczynska
- Department and Clinic of Orthopaedics and Traumatology, Medical University of Lublin, Jaczewskiego 8 Street, 20-090 Lublin, Poland; (M.T.); (K.G.)
- Arthros Medical Centre, Chodzki 31 Street, 20-093 Lublin, Poland
| | - Krzysztof Gaweda
- Department and Clinic of Orthopaedics and Traumatology, Medical University of Lublin, Jaczewskiego 8 Street, 20-090 Lublin, Poland; (M.T.); (K.G.)
- Arthros Medical Centre, Chodzki 31 Street, 20-093 Lublin, Poland
| | - Katarzyna Klimek
- Chair and Department of Biochemistry and Biotechnology, Medical University of Lublin, Chodzki Street 1, 20-093 Lublin, Poland; (A.G.); (A.H.)
| |
Collapse
|
3
|
Sargenti A, Pasqua S, Leu M, Dionisi L, Filardo G, Grigolo B, Gazzola D, Santi S, Cavallo C. Adipose Stromal Cell Spheroids for Cartilage Repair: A Promising Tool for Unveiling the Critical Maturation Point. Bioengineering (Basel) 2023; 10:1182. [PMID: 37892912 PMCID: PMC10603958 DOI: 10.3390/bioengineering10101182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 10/09/2023] [Accepted: 10/10/2023] [Indexed: 10/29/2023] Open
Abstract
Articular cartilage lacks intrinsic regenerative capabilities, and the current treatments fail to regenerate damaged tissue and lead only to temporary pain relief. These limitations have prompted the development of tissue engineering approaches, including 3D culture systems. Thanks to their regenerative properties and capacity to recapitulate embryonic processes, spheroids obtained from mesenchymal stromal cells are increasingly studied as building blocks to obtain functional tissues. The aim of this study was to investigate the capacity of adipose stromal cells to assemble in spheroids and differentiate toward chondrogenic lineage from the perspective of cartilage repair. Spheroids were generated by two different methods (3D chips vs. Ultra-Low Attachment plates), differentiated towards chondrogenic lineage, and their properties were investigated using molecular biology analyses, biophysical measurement of mass density, weight, and size of spheroids, and confocal imaging. Overall, spheroids showed the ability to differentiate by expressing specific cartilaginous markers that correlate with their mass density, defining a critical point at which they start to mature. Considering the spheroid generation method, this pilot study suggested that spheroids obtained with chips are a promising tool for the generation of cartilage organoids that could be used for preclinical/clinical approaches, including personalized therapy.
Collapse
Affiliation(s)
- Azzurra Sargenti
- CellDynamics iSRL, 40136 Bologna, Italy; (A.S.); (S.P.); (L.D.); (D.G.)
| | - Simone Pasqua
- CellDynamics iSRL, 40136 Bologna, Italy; (A.S.); (S.P.); (L.D.); (D.G.)
| | - Marco Leu
- abc biopply ag, 4500 Solothurn, Switzerland;
| | - Laura Dionisi
- CellDynamics iSRL, 40136 Bologna, Italy; (A.S.); (S.P.); (L.D.); (D.G.)
| | - Giuseppe Filardo
- Applied and Translational Research (ATR) Center, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy;
| | - Brunella Grigolo
- Laboratorio RAMSES, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy;
| | - Daniele Gazzola
- CellDynamics iSRL, 40136 Bologna, Italy; (A.S.); (S.P.); (L.D.); (D.G.)
| | - Spartaco Santi
- Institute of Molecular Genetics “Luigi Luca Cavalli-Sforza”, Unit of Bologna, CNR, 40136 Bologna, Italy
- IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy
| | - Carola Cavallo
- Laboratorio RAMSES, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy;
| |
Collapse
|
4
|
Vaca-González JJ, Culma JJS, Nova LMH, Garzón-Alvarado DA. Anatomy, molecular structures, and hyaluronic acid - Gelatin injectable hydrogels as a therapeutic alternative for hyaline cartilage recovery: A review. J Biomed Mater Res B Appl Biomater 2023. [PMID: 37178328 DOI: 10.1002/jbm.b.35261] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 04/24/2023] [Accepted: 05/03/2023] [Indexed: 05/15/2023]
Abstract
Cartilage damage caused by trauma or osteoarthritis is a common joint disease that can increase the social and economic burden in society. Due to its avascular characteristics, the poor migration ability of chondrocytes, and a low number of progenitor cells, the self-healing ability of cartilage defects has been significantly limited. Hydrogels have been developed into one of the most suitable biomaterials for the regeneration of cartilage because of its characteristics such as high-water absorption, biodegradation, porosity, and biocompatibility similar to natural extracellular matrix. Therefore, the present review article presents a conceptual framework that summarizes the anatomical, molecular structure and biochemical properties of hyaline cartilage located in long bones: articular cartilage and growth plate. Moreover, the importance of preparation and application of hyaluronic acid - gelatin hydrogels for cartilage tissue engineering are included. Hydrogels possess benefits of stimulating the production of Agc1, Col2α1-IIa, and SOX9, molecules important for the synthesis and composition of the extracellular matrix of cartilage. Accordingly, they are believed to be promising biomaterials of therapeutic alternatives to treat cartilage damage.
Collapse
Affiliation(s)
- Juan Jairo Vaca-González
- Escuela de Pregrado, Dirección Académica, Vicerrectoría de Sede, Universidad Nacional de Colombia, Sede de La Paz, Cesar, Colombia
- Biomimetics Laboratory, Biotechnology Institute, Universidad Nacional de Colombia, Bogotá, Colombia
| | - Juan José Saiz Culma
- Biomimetics Laboratory, Biotechnology Institute, Universidad Nacional de Colombia, Bogotá, Colombia
| | | | - Diego Alexander Garzón-Alvarado
- Biomimetics Laboratory, Biotechnology Institute, Universidad Nacional de Colombia, Bogotá, Colombia
- Numerical Methods and Modeling Research Group (GNUM), Universidad Nacional de Colombia, Bogotá, Colombia
| |
Collapse
|
5
|
Kilian D, Poddar A, Desrochers V, Heinemann C, Halfter N, Liu S, Rother S, Gelinsky M, Hintze V, Lode A. Cellular adhesion and chondrogenic differentiation inside an alginate-based bioink in response to tailorable artificial matrices and tannic acid treatment. BIOMATERIALS ADVANCES 2023; 147:213319. [PMID: 36758282 DOI: 10.1016/j.bioadv.2023.213319] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 12/30/2022] [Accepted: 01/27/2023] [Indexed: 02/04/2023]
Abstract
Many established bioinks fulfill important requirements regarding fabrication standards and cytocompatibility. Current research focuses on development of functionalized bioinks with an improved support of tissue-specific cell differentiation. Many approaches primarily depend on decellularized extracellular matrices or blood components. In this study, we investigated the combination of a highly viscous alginate-methylcellulose (algMC) bioink with collagen-based artificial extracellular matrix (aECM) as a finely controllable and tailorable system composed of collagen type I (col) with and without chondroitin sulfate (CS) or sulfated hyaluronan (sHA). As an additional stabilizer, the polyphenol tannic acid (TA) was integrated into the inks. The assessment of rheological properties and printability as well as hydrogel microstructure revealed no adverse effect of the integrated components on the inks. Viability, adhesion, and proliferation of bioprinted immortalized human mesenchymal stem cells (hTERT-MSC) was improved indicating enhanced interaction with the designed microenvironment. Furthermore, chondrogenic matrix production (collagen type II and sulfated glycosaminoglycans) by primary human chondrocytes (hChon) was enhanced by aECM. Supplementing the inks with TA was required for these positive effects but caused cytotoxicity as soon as TA concentrations exceeded a certain amount. Thus, combining tailorable aECM with algMC and balanced TA addition proved to be a promising approach for promoting adhesion of immortalized stem cells and differentiation of chondrocytes in bioprinted scaffolds.
Collapse
Affiliation(s)
- David Kilian
- Centre for Translational Bone, Joint and Soft Tissue Research, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Fetscherstrasse 74, 01307 Dresden, Germany
| | - Aayush Poddar
- Centre for Translational Bone, Joint and Soft Tissue Research, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Fetscherstrasse 74, 01307 Dresden, Germany
| | - Vanessa Desrochers
- Centre for Translational Bone, Joint and Soft Tissue Research, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Fetscherstrasse 74, 01307 Dresden, Germany
| | - Christiane Heinemann
- Institute of Materials Science, Max Bergmann Center of Biomaterials, Technische Universität Dresden, Budapester Strasse 27, 01069 Dresden, Germany
| | - Norbert Halfter
- Institute of Materials Science, Max Bergmann Center of Biomaterials, Technische Universität Dresden, Budapester Strasse 27, 01069 Dresden, Germany
| | - Suihong Liu
- Centre for Translational Bone, Joint and Soft Tissue Research, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Fetscherstrasse 74, 01307 Dresden, Germany; Rapid Manufacturing Engineering Center, School of Mechatronic Engineering and Automation, Shanghai University, Shanghai 200444, China
| | - Sandra Rother
- Institute of Materials Science, Max Bergmann Center of Biomaterials, Technische Universität Dresden, Budapester Strasse 27, 01069 Dresden, Germany; Center for Molecular Signaling (PZMS), Saarland University School of Medicine, Homburg, Saar, Germany
| | - Michael Gelinsky
- Centre for Translational Bone, Joint and Soft Tissue Research, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Fetscherstrasse 74, 01307 Dresden, Germany
| | - Vera Hintze
- Institute of Materials Science, Max Bergmann Center of Biomaterials, Technische Universität Dresden, Budapester Strasse 27, 01069 Dresden, Germany
| | - Anja Lode
- Centre for Translational Bone, Joint and Soft Tissue Research, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Fetscherstrasse 74, 01307 Dresden, Germany.
| |
Collapse
|
6
|
Talouki PY, Tackallou SH, Shojaei S, Benisi SZ, Goodarzi V. The role of three-dimensional scaffolds based on polyglycerol sebacate/ polycaprolactone/ gelatin in the presence of Nanohydroxyapatite in promoting chondrogenic differentiation of human adipose-derived mesenchymal stem cells. Biol Proced Online 2023; 25:9. [PMID: 36964481 PMCID: PMC10039520 DOI: 10.1186/s12575-023-00197-z] [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: 01/11/2023] [Accepted: 03/18/2023] [Indexed: 03/26/2023] Open
Abstract
BACKGROUND Tissue engineering for cartilage regeneration has made great advances in recent years, although there are still challenges to overcome. This study aimed to evaluate the chondrogenic differentiation of human adipose-derived mesenchymal stem cells (hADSCs) on three-dimensional scaffolds based on polyglycerol sebacate (PGS) / polycaprolactone (PCL) / gelatin(Gel) in the presence of Nanohydroxyapatite (nHA). MATERIALS AND METHODS In this study, a series of nHA-nanocomposite scaffolds were fabricated using 100:0:0, 60:40:0, and 60:20:20 weight ratios of PGS to PCL: Gel copolymers through salt leaching method. The morphology and porosity of prepared samples was characterized by SEM and EDX mapping analysis. Also, the dynamic contact angle and PBS adsorption tests are used to identify the effect of copolymerization and nanoparticles on scaffolds' hydrophilicity. The hydrolytic degradation properties were also analyzed. Furthermore, cell viability and proliferation as well as cell adhesion are evaluated to find out the biocompatibility. To determine the potential ability of nHA-nanocomposite scaffolds in chondrogenic differentiation, RT-PCR assay was performed to monitor the expression of collagen II, aggrecan, and Sox9 genes as markers of cartilage differentiation. RESULTS The nanocomposites had an elastic modulus within a range of 0.71-1.30 MPa and 0.65-0.43 MPa, in dry and wet states, respectively. The PGS/PCL sample showed a water contact angle of 72.44 ± 2.2°, while the hydrophilicity significantly improved by adding HA nanoparticles. It was found from the hydrolytic degradation study that HA incorporation can accelerate the degradation rate compared with PGS and PGS/PCL samples. Furthermore, the in vitro biocompatibility tests showed significant cell attachment, proliferation, and viability of adipose-derived mesenchymal stem cells (ADMSCs). RT-PCR also indicated a significant increase in collagen II, aggrecan and Sox9 mRNA levels. CONCLUSIONS Our findings demonstrated that these nanocomposite scaffolds promote the differentiation of hADSCs into chondrocytes possibly by the increase in mRNA levels of collagen II, aggrecan, and Sox9 as markers of chondrogenic differentiation. In conclusion, the addition of PCL, Gelatin, and HA into PGS is a practical approach to adjust the general features of PGS to prepare a promising scaffold for cartilage tissue engineering.
Collapse
Affiliation(s)
- Pardis Yousefi Talouki
- Department of Biomedical Engineering, Central Branch, Islamic Azad University, Tehran, Iran
| | - Saeed Hesami Tackallou
- Department of Biology, Central Branch, Islamic Azad University, P.O. Box 13145-784, Tehran, Iran.
| | - Shahrokh Shojaei
- Department of Biomedical Engineering, Central Branch, Islamic Azad University, Tehran, Iran
- Stem Cell Research Center, Tissue Engineering and Regenerative Medicine Institute, Islamic Azad University, Central Branch, Tehran, Iran
| | - Soheila Zamanlui Benisi
- Department of Biomedical Engineering, Central Branch, Islamic Azad University, Tehran, Iran
- Stem Cell Research Center, Tissue Engineering and Regenerative Medicine Institute, Islamic Azad University, Central Branch, Tehran, Iran
| | - Vahabodin Goodarzi
- Applied Biotechnology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| |
Collapse
|
7
|
Majood M, Shakeel A, Agarwal A, Jeevanandham S, Bhattacharya R, Kochhar D, Singh A, Kalyanasundaram D, Mohanty S, Mukherjee M. Hydrogel Nanosheets Confined 2D Rhombic Ice: A New Platform Enhancing Chondrogenesis. Biomed Mater 2022; 17. [PMID: 36044885 DOI: 10.1088/1748-605x/ac8e43] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 08/31/2022] [Indexed: 11/12/2022]
Abstract
Nanoconfinement within flexible interfaces is a key step towards exploiting confinement effects in several biological and technological systems wherein flexible 2D materials are frequently utilized but are arduous to prepare. Hitherto unreported, the synthesis of 2D Hydrogel nanosheets (HNS) using a template- and catalyst-free process is developed representing a fertile ground for fundamental structure-property investigations. In due course of time, nucleating folds propagating along the edges trigger co-operative deformations of HNS generating regions of nanoconfinement within trapped water islands. These severely constricting surfaces force water molecules to pack within the nanoscale regime of HNS almost parallel to the surface bringing about phase transition into puckered rhombic ice with AA and AB Bernal stacking pattern, which was mostly restricted to Molecular dynamics (MD) studies so far. Interestingly, under high lateral pressure and spatial inhomogeneity within nanoscale confinement, bilayer rhombic ice structures were formed with an in-plane lattice spacing of 0.31 nm. In this work, a systematic exploration of rhombic ice formation within HNS has been delineated using High-resolution transmission electron microscopy (HRTEM), and its ultrathin morphology was examined using Atomic Force Microscopy (AFM). Scanning Electron Microscopy (SEM) images revealed high porosity while mechanical testing presented young's modulus of 155 kPa with ~84% deformation, whereas contact angle suggested high hydrophilicity. The combinations of nanosheets, porosity, nanoconfinement, hydrophilicity, and mechanical strength, motivated us to explore their application as a scaffold for cartilage regeneration, by inducing chondrogenesis of human Wharton Jelly derived mesenchymal stem cells (hWJ MSCs). HNS promoted the formation of cell aggregates giving higher number of spheroid formation and a marked expression of chondrogenic markers (ColI, ColII, ColX, ACAN and S-100), thereby providing some cues for guiding chondrogenic differentiation.
Collapse
Affiliation(s)
- Misba Majood
- AICCRS, Amity University, Sector 125, Noida, Noida, Uttar Pradesh, 201313, INDIA
| | - Adeeba Shakeel
- AICCRS, Amity University, Sector 125, Noida, Uttar Pradesh, 201313, INDIA
| | - Aakanksha Agarwal
- AICCRS, Amity University, Sector 125, Noida, Uttar Pradesh, 201313, INDIA
| | | | | | - Dakshi Kochhar
- Amity University, Sector 125, Noida, Uttar Pradesh, 201313, INDIA
| | - Aarti Singh
- AICCRS, Amity University, Sector 125, Noida, Uttar Pradesh, 201313, INDIA
| | | | - Sujata Mohanty
- Stem Cell Facility, All India Institute of Medical Sciences Cardio-Thoracic Sciences Centre, Orbo Building, first floor,, Ansari Nagar, New Delhi, New Delhi, Delhi, 110029, INDIA
| | | |
Collapse
|
8
|
Rizzo MG, Palermo N, D’Amora U, Oddo S, Guglielmino SPP, Conoci S, Szychlinska MA, Calabrese G. Multipotential Role of Growth Factor Mimetic Peptides for Osteochondral Tissue Engineering. Int J Mol Sci 2022; 23:ijms23137388. [PMID: 35806393 PMCID: PMC9266819 DOI: 10.3390/ijms23137388] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 06/28/2022] [Accepted: 06/30/2022] [Indexed: 12/25/2022] Open
Abstract
Articular cartilage is characterized by a poor self-healing capacity due to its aneural and avascular nature. Once injured, it undergoes a series of catabolic processes which lead to its progressive degeneration and the onset of a severe chronic disease called osteoarthritis (OA). In OA, important alterations of the morpho-functional organization occur in the cartilage extracellular matrix, involving all the nearby tissues, including the subchondral bone. Osteochondral engineering, based on a perfect combination of cells, biomaterials and biomolecules, is becoming increasingly successful for the regeneration of injured cartilage and underlying subchondral bone tissue. To this end, recently, several peptides have been explored as active molecules and enrichment motifs for the functionalization of biomaterials due to their ability to be easily chemically synthesized, as well as their tunable physico-chemical features, low immunogenicity issues and functional group modeling properties. In addition, they have shown a good aptitude to penetrate into the tissue due to their small size and stability at room temperature. In particular, growth-factor-derived peptides can play multiple functions in bone and cartilage repair, exhibiting chondrogenic/osteogenic differentiation properties. Among the most studied peptides, great attention has been paid to transforming growth factor-β and bone morphogenetic protein mimetic peptides, cell-penetrating peptides, cell-binding peptides, self-assembling peptides and extracellular matrix-derived peptides. Moreover, recently, phage display technology is emerging as a powerful selection technique for obtaining functional peptides on a large scale and at a low cost. In particular, these peptides have demonstrated advantages such as high biocompatibility; the ability to be immobilized directly on chondro- and osteoinductive nanomaterials; and improving the cell attachment, differentiation, development and regeneration of osteochondral tissue. In this context, the aim of the present review was to go through the recent literature underlining the importance of studying novel functional motifs related to growth factor mimetic peptides that could be a useful tool in osteochondral repair strategies. Moreover, the review summarizes the current knowledge of the use of phage display peptides in osteochondral tissue regeneration.
Collapse
Affiliation(s)
- Maria Giovanna Rizzo
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno d’Alcontres, 31, 98168 Messina, Italy; (M.G.R.); (N.P.); (S.O.); (S.P.P.G.)
| | - Nicoletta Palermo
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno d’Alcontres, 31, 98168 Messina, Italy; (M.G.R.); (N.P.); (S.O.); (S.P.P.G.)
| | - Ugo D’Amora
- Institute of Polymers, Composites and Biomaterials—National Research Council, Viale J. F. Kennedy 54, Mostra d’Oltremare, Pad. 20, 80125 Naples, Italy;
| | - Salvatore Oddo
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno d’Alcontres, 31, 98168 Messina, Italy; (M.G.R.); (N.P.); (S.O.); (S.P.P.G.)
| | - Salvatore Pietro Paolo Guglielmino
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno d’Alcontres, 31, 98168 Messina, Italy; (M.G.R.); (N.P.); (S.O.); (S.P.P.G.)
| | - Sabrina Conoci
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno d’Alcontres, 31, 98168 Messina, Italy; (M.G.R.); (N.P.); (S.O.); (S.P.P.G.)
- Department of Chemistry “Giacomo Ciamician”, University of Bologna, Via Selmi 2, 40126 Bologna, Italy
- Correspondence: (S.C.); (G.C.)
| | - Marta Anna Szychlinska
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (Bi.N.D.), University of Palermo, Via del Vespro, 129, 90127 Palermo, Italy;
| | - Giovanna Calabrese
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno d’Alcontres, 31, 98168 Messina, Italy; (M.G.R.); (N.P.); (S.O.); (S.P.P.G.)
- Correspondence: (S.C.); (G.C.)
| |
Collapse
|
9
|
Zheng J, Xie Y, Yoshitomi T, Kawazoe N, Yang Y, Chen G. Stepwise Proliferation and Chondrogenic Differentiation of Mesenchymal Stem Cells in Collagen Sponges under Different Microenvironments. Int J Mol Sci 2022; 23:ijms23126406. [PMID: 35742851 PMCID: PMC9223568 DOI: 10.3390/ijms23126406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 06/03/2022] [Accepted: 06/06/2022] [Indexed: 01/27/2023] Open
Abstract
Biomimetic microenvironments are important for controlling stem cell functions. In this study, different microenvironmental conditions were investigated for the stepwise control of proliferation and chondrogenic differentiation of human bone-marrow-derived mesenchymal stem cells (hMSCs). The hMSCs were first cultured in collagen porous sponges and then embedded with or without collagen hydrogels for continual culture under different culture conditions. The different influences of collagen sponges, collagen hydrogels, and induction factors were investigated. The collagen sponges were beneficial for cell proliferation. The collagen sponges also promoted chondrogenic differentiation during culture in chondrogenic medium, which was superior to the effect of collagen sponges embedded with hydrogels without loading of induction factors. However, collagen sponges embedded with collagen hydrogels and loaded with induction factors had the same level of promotive effect on chondrogenic differentiation as collagen sponges during in vitro culture in chondrogenic medium and showed the highest promotive effect during in vivo subcutaneous implantation. The combination of collagen sponges with collagen hydrogels and induction factors could provide a platform for cell proliferation at an early stage and subsequent chondrogenic differentiation at a late stage. The results provide useful information for the chondrogenic differentiation of stem cells and cartilage tissue engineering.
Collapse
Affiliation(s)
- Jing Zheng
- Research Center for Functional Materials, National Institute for Materials Science, 1-1 Namiki, Tsukuba, Ibaraki 305-0044, Japan; (J.Z.); (Y.X.); (T.Y.); (N.K.)
- Department of Materials Science and Engineering, Graduate School of Pure and Applied Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8577, Japan
| | - Yan Xie
- Research Center for Functional Materials, National Institute for Materials Science, 1-1 Namiki, Tsukuba, Ibaraki 305-0044, Japan; (J.Z.); (Y.X.); (T.Y.); (N.K.)
- Department of Materials Science and Engineering, Graduate School of Pure and Applied Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8577, Japan
| | - Toru Yoshitomi
- Research Center for Functional Materials, National Institute for Materials Science, 1-1 Namiki, Tsukuba, Ibaraki 305-0044, Japan; (J.Z.); (Y.X.); (T.Y.); (N.K.)
| | - Naoki Kawazoe
- Research Center for Functional Materials, National Institute for Materials Science, 1-1 Namiki, Tsukuba, Ibaraki 305-0044, Japan; (J.Z.); (Y.X.); (T.Y.); (N.K.)
| | - Yingnan Yang
- Graduate School of Life and Environmental Science, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8572, Japan;
| | - Guoping Chen
- Research Center for Functional Materials, National Institute for Materials Science, 1-1 Namiki, Tsukuba, Ibaraki 305-0044, Japan; (J.Z.); (Y.X.); (T.Y.); (N.K.)
- Department of Materials Science and Engineering, Graduate School of Pure and Applied Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8577, Japan
- Correspondence: ; Tel.: +81-29-860-4496
| |
Collapse
|
10
|
Shao R, Dong Y, Zhang S, Wu X, Huang X, Sun B, Zeng B, Xu F, Liang W. State of the art of bone biomaterials and their interactions with stem cells: Current state and future directions. Biotechnol J 2022; 17:e2100074. [PMID: 35073451 DOI: 10.1002/biot.202100074] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 12/07/2021] [Accepted: 12/08/2021] [Indexed: 11/11/2022]
Affiliation(s)
- Ruyi Shao
- Department of Orthopedics Zhuji People's Hospital Shaoxing Zhejiang Province 312500 P. R. China
| | - Yongqiang Dong
- Department of Orthopaedics Xinchang People's Hospital Shaoxing Zhejiang Province 312500 P. R. China
| | - Songou Zhang
- College of Medicine Shaoxing University Shaoxing Zhejiang Province 312000 P. R. China
| | - Xudong Wu
- Department of Orthopedics Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University 355 Xinqiao Road, Dinghai District Zhoushan Zhejiang Province 316000 P. R. China
| | - Xiaogang Huang
- Department of Orthopedics Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University 355 Xinqiao Road, Dinghai District Zhoushan Zhejiang Province 316000 P. R. China
| | - Bin Sun
- Department of Orthopedics Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University 355 Xinqiao Road, Dinghai District Zhoushan Zhejiang Province 316000 P. R. China
| | - Bin Zeng
- Department of Orthopedics Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University 355 Xinqiao Road, Dinghai District Zhoushan Zhejiang Province 316000 P. R. China
| | - Fangming Xu
- Department of Orthopedics Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University 355 Xinqiao Road, Dinghai District Zhoushan Zhejiang Province 316000 P. R. China
| | - Wenqing Liang
- Department of Orthopedics Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University 355 Xinqiao Road, Dinghai District Zhoushan Zhejiang Province 316000 P. R. China
| |
Collapse
|
11
|
Roncada T, Bonithon R, Blunn G, Roldo M. Soft substrates direct stem cell differentiation into the chondrogenic lineage without the use of growth factors. J Tissue Eng 2022; 13:20417314221122121. [PMID: 36199979 PMCID: PMC9528007 DOI: 10.1177/20417314221122121] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 08/03/2022] [Indexed: 12/03/2022] Open
Abstract
Mesenchymal stem cells (MSCs) hold great promise for the treatment of cartilage related injuries. However, selectively promoting stem cell differentiation in vivo is still challenging. Chondrogenic differentiation of MSCs usually requires the use of growth factors that lead to the overexpression of hypertrophic markers. In this study, for the first time the effect of stiffness on MSC differentiation has been tested without the use of growth factors. Three-dimensional collagen and alginate scaffolds were developed and characterised. Stiffness significantly affected gene expression and ECM deposition. While, all hydrogels supported chondrogenic differentiation and allowed deposition of collagen type II and aggrecan, the 5.75 kPa hydrogel showed limited production of collagen type I compared to the other two formulations. These findings demonstrated for the first time that stiffness can guide MSCs differentiation without the use of growth factors within a tissue engineering scaffold suitable for the treatment of cartilage defects.
Collapse
Affiliation(s)
- Tosca Roncada
- School of Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth, UK
| | - Roxane Bonithon
- School of Mechanical and Design Engineering, University of Portsmouth, Portsmouth, UK
| | - Gordon Blunn
- School of Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth, UK
| | - Marta Roldo
- School of Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth, UK
- Marta Roldo, School of Pharmacy and Biomedical Sciences, University of Portsmouth, White Swan Road, Portsmouth, PO1 2DT, UK.
| |
Collapse
|
12
|
Shestovskaya MV, Bozhkova SA, Sopova JV, Khotin MG, Bozhokin MS. Methods of Modification of Mesenchymal Stem Cells and Conditions of Their Culturing for Hyaline Cartilage Tissue Engineering. Biomedicines 2021; 9:biomedicines9111666. [PMID: 34829895 PMCID: PMC8615732 DOI: 10.3390/biomedicines9111666] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 11/04/2021] [Accepted: 11/05/2021] [Indexed: 12/24/2022] Open
Abstract
The use of mesenchymal stromal cells (MSCs) for tissue engineering of hyaline cartilage is a topical area of regenerative medicine that has already entered clinical practice. The key stage of this procedure is to create conditions for chondrogenic differentiation of MSCs, increase the synthesis of hyaline cartilage extracellular matrix proteins by these cells and activate their proliferation. The first such works consisted in the indirect modification of cells, namely, in changing the conditions in which they are located, including microfracturing of the subchondral bone and the use of 3D biodegradable scaffolds. The most effective methods for modifying the cell culture of MSCs are protein and physical, which have already been partially introduced into clinical practice. Genetic methods for modifying MSCs, despite their effectiveness, have significant limitations. Techniques have not yet been developed that allow studying the effectiveness of their application even in limited groups of patients. The use of MSC modification methods allows precise regulation of cell culture proliferation, and in combination with the use of a 3D biodegradable scaffold, it allows obtaining a hyaline-like regenerate in the damaged area. This review is devoted to the consideration and comparison of various methods used to modify the cell culture of MSCs for their use in regenerative medicine of cartilage tissue.
Collapse
Affiliation(s)
- Maria V. Shestovskaya
- Institute of Cytology of the Russian Academy of Sciences, Tikhoretsky Ave. 4, 194064 St. Petersburg, Russia; (M.V.S.); (J.V.S.); (M.G.K.)
| | - Svetlana A. Bozhkova
- Vreden National Medical Research Center of Traumatology and Orthopedics, Academica Baykova Str., 8, 195427 St. Petersburg, Russia;
| | - Julia V. Sopova
- Institute of Cytology of the Russian Academy of Sciences, Tikhoretsky Ave. 4, 194064 St. Petersburg, Russia; (M.V.S.); (J.V.S.); (M.G.K.)
- Center of Transgenesis and Genome Editing, St. Petersburg State University, Universitetskaja Emb., 7/9, 199034 St. Petersburg, Russia
| | - Mikhail G. Khotin
- Institute of Cytology of the Russian Academy of Sciences, Tikhoretsky Ave. 4, 194064 St. Petersburg, Russia; (M.V.S.); (J.V.S.); (M.G.K.)
| | - Mikhail S. Bozhokin
- Institute of Cytology of the Russian Academy of Sciences, Tikhoretsky Ave. 4, 194064 St. Petersburg, Russia; (M.V.S.); (J.V.S.); (M.G.K.)
- Vreden National Medical Research Center of Traumatology and Orthopedics, Academica Baykova Str., 8, 195427 St. Petersburg, Russia;
- Correspondence:
| |
Collapse
|
13
|
Huynh PD, Vu NB, To XHV, Le TM. Culture and Differentiation of Human Umbilical Cord-Derived Mesenchymal Stem Cells on Growth Factor-Rich Fibrin Scaffolds to Produce Engineered Cartilages. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021:193-208. [PMID: 34739721 DOI: 10.1007/5584_2021_670] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
INTRODUCTION After injuries, the cartilage healing capacity is limited owing to its nature as a particular connective tissue without blood vessels, lymphatics, or nerves. The creation of artificial cartilage tissue mimics the biological properties of native cartilage and can reduce the need for donated tissue. Fibrin is a type of biodegradable scaffold that has great potential in tissue engineering applications. It can become good material for cell adhesion and proliferation in vitro. Therefore, this study aimed to create a cartilage tissue in vitro using umbilical cord-derived mesenchymal stem cells (UCMSC) and growth factor-rich fibrin (GRF) scaffolds. METHODS UCMSCs were isolated and expanded, and platelet-rich plasma (PRP) preparations were performed following previously published protocols. PRP was activated (aPRP) by a 0.45-μm syringe filter to release growth factors inside the platelets. Each 2.105 of the UCMSCs were suspended in 2 ml of aPRP to make the mixture of MSC and PRP (MSC-PRP). Then, Ca2+ solution was added to this mixture to produce the fibril scaffold with UCMSCs inside. UCMSCs' adhesion and proliferation inside the scaffold were evaluated by observation under inverted microscopy, H-E staining, MTT assays, and scanning electron microscopy (SEM). The fibril structure containing UCMSCs was cultured, and chondrogenesis was induced using commercial chondrogenesis media for 21 days (iMSC-GRF). The differentiation in efficacy toward cartilage was evaluated based on the accumulation of aggrecan (acan), glycosaminoglycans (GAGs), and collagen type II (Col II). RESULTS The results showed that we successfully created a cartilage tissue with some characteristics that mimic the properties of natural cartilage. The engineered cartilage tissue was positive with some cartilage protein, such as acan, GAG, and Coll II. In vitro cartilage presented some natural chondrocyte-like cells. The artificial cartilage tissue was positive for CD14, CD34, CD90, CD105, and HLA-DR and negative for CD44, CD45, and CD73. CONCLUSION These results showed that using UCMSCs and growth factor-rich fibril from platelet-rich plasma was feasible to produce engineered cartilage tissue for further experiments or clinical usage.
Collapse
Affiliation(s)
- Phat Duc Huynh
- Laboratory of Stem Cell Research and Application, University of Science Ho Chi Minh City, Ho Chi Minh City, Vietnam
- Vietnam National University Ho Chi Minh City, Ho Chi Minh City, Vietnam
| | - Ngoc Bich Vu
- Vietnam National University Ho Chi Minh City, Ho Chi Minh City, Vietnam.
- Stem Cell Institute, University of Science Ho Chi Minh City, Ho Chi Minh City, Vietnam.
| | - Xuan Hoang-Viet To
- Vietnam National University Ho Chi Minh City, Ho Chi Minh City, Vietnam
- Stem Cell Institute, University of Science Ho Chi Minh City, Ho Chi Minh City, Vietnam
| | - Thuan Minh Le
- Laboratory of Stem Cell Research and Application, University of Science Ho Chi Minh City, Ho Chi Minh City, Vietnam
- Vietnam National University Ho Chi Minh City, Ho Chi Minh City, Vietnam
| |
Collapse
|
14
|
Sawadkar P, Mandakhbayar N, Patel KD, Buitrago JO, Kim TH, Rajasekar P, Lali F, Kyriakidis C, Rahmani B, Mohanakrishnan J, Dua R, Greco K, Lee JH, Kim HW, Knowles J, García-Gareta E. Three dimensional porous scaffolds derived from collagen, elastin and fibrin proteins orchestrate adipose tissue regeneration. J Tissue Eng 2021; 12:20417314211019238. [PMID: 34104389 PMCID: PMC8165536 DOI: 10.1177/20417314211019238] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 05/04/2021] [Indexed: 12/18/2022] Open
Abstract
Current gold standard to treat soft tissue injuries caused by trauma and pathological condition are autografts and off the shelf fillers, but they have inherent weaknesses like donor site morbidity, immuno-compatibility and graft failure. To overcome these limitations, tissue-engineered polymers are seeded with stem cells to improve the potential to restore tissue function. However, their interaction with native tissue is poorly understood so far. To study these interactions and improve outcomes, we have fabricated scaffolds from natural polymers (collagen, fibrin and elastin) by custom-designed processes and their material properties such as surface morphology, swelling, wettability and chemical cross-linking ability were characterised. By using 3D scaffolds, we comprehensive assessed survival, proliferation and phenotype of adipose-derived stem cells in vitro. In vivo, scaffolds were seeded with adipose-derived stem cells and implanted in a rodent model, with X-ray microtomography, histology and immunohistochemistry as read-outs. Collagen-based materials showed higher cell adhesion and proliferation in vitro as well as higher adipogenic properties in vivo. In contrast, fibrin demonstrated poor cellular and adipogenesis properties but higher angiogenesis. Elastin formed the most porous scaffold, with cells displaying a non-aggregated morphology in vitro while in vivo elastin was the most degraded scaffold. These findings of how polymers present in the natural polymers mimicking ECM and seeded with stem cells affect adipogenesis in vitro and in vivo can open avenues to design 3D grafts for soft tissue repair.
Collapse
Affiliation(s)
- Prasad Sawadkar
- Regenerative Biomaterials Group, The RAFT Institute and The Griffin Institute, Northwick Park & Saint Mark's Hospital, London, UK.,Division of Surgery and Interventional Science, University College London, London, UK.,UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, Cheonan, Republic of Korea
| | - Nandin Mandakhbayar
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, Republic of Korea.,Department of Nanobiomedical Science & BK21 Plus NBM Global Research Centre for Regenerative Medicine, Dankook University, Cheonan, Republic of Korea.,Department of Biomaterials Science, School of Dentistry, Dankook University, Cheonan, Republic of Korea
| | - Kapil D Patel
- UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, Cheonan, Republic of Korea.,Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, Republic of Korea.,Department of Nanobiomedical Science & BK21 Plus NBM Global Research Centre for Regenerative Medicine, Dankook University, Cheonan, Republic of Korea.,Division of Biomaterials and Tissue Engineering, UCL Eastman Dental Institute, University College London, London, UK
| | - Jennifer Olmas Buitrago
- UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, Cheonan, Republic of Korea.,Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, Republic of Korea.,Department of Nanobiomedical Science & BK21 Plus NBM Global Research Centre for Regenerative Medicine, Dankook University, Cheonan, Republic of Korea
| | - Tae Hyun Kim
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, Republic of Korea.,Department of Nanobiomedical Science & BK21 Plus NBM Global Research Centre for Regenerative Medicine, Dankook University, Cheonan, Republic of Korea.,R&D Center, TE Bios Co, Osong, Republic of Korea
| | - Poojitha Rajasekar
- Division of Respiratory Medicine, University of Nottingham, Nottingham, UK
| | - Ferdinand Lali
- Division of Surgery and Interventional Science, University College London, London, UK.,The Griffin Institute, Northwick Park and St Mark's Hospital, London, UK
| | - Christos Kyriakidis
- Regenerative Biomaterials Group, The RAFT Institute and The Griffin Institute, Northwick Park & Saint Mark's Hospital, London, UK
| | - Benyamin Rahmani
- Department of Mechanical Engineering, University College London, London, UK
| | - Jeviya Mohanakrishnan
- Regenerative Biomaterials Group, The RAFT Institute and The Griffin Institute, Northwick Park & Saint Mark's Hospital, London, UK
| | - Rishbha Dua
- Regenerative Biomaterials Group, The RAFT Institute and The Griffin Institute, Northwick Park & Saint Mark's Hospital, London, UK
| | - Karin Greco
- Division of Surgery and Interventional Science, University College London, London, UK.,The Griffin Institute, Northwick Park and St Mark's Hospital, London, UK
| | - Jung-Hwan Lee
- UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, Cheonan, Republic of Korea.,Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, Republic of Korea.,Department of Nanobiomedical Science & BK21 Plus NBM Global Research Centre for Regenerative Medicine, Dankook University, Cheonan, Republic of Korea.,Department of Biomaterials Science, School of Dentistry, Dankook University, Cheonan, Republic of Korea
| | - Hae-Won Kim
- UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, Cheonan, Republic of Korea.,Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, Republic of Korea.,Department of Nanobiomedical Science & BK21 Plus NBM Global Research Centre for Regenerative Medicine, Dankook University, Cheonan, Republic of Korea.,Department of Biomaterials Science, School of Dentistry, Dankook University, Cheonan, Republic of Korea
| | - Jonathan Knowles
- UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, Cheonan, Republic of Korea.,Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, Republic of Korea.,Department of Nanobiomedical Science & BK21 Plus NBM Global Research Centre for Regenerative Medicine, Dankook University, Cheonan, Republic of Korea.,Department of Biomaterials Science, School of Dentistry, Dankook University, Cheonan, Republic of Korea.,Division of Biomaterials and Tissue Engineering, UCL Eastman Dental Institute, University College London, London, UK
| | - Elena García-Gareta
- Regenerative Biomaterials Group, The RAFT Institute and The Griffin Institute, Northwick Park & Saint Mark's Hospital, London, UK.,Division of Biomaterials and Tissue Engineering, UCL Eastman Dental Institute, University College London, London, UK
| |
Collapse
|
15
|
Ye M, Yu L, She Y, Wang S, Wang M, Zhao Q, Gu C, Bian L, Wen N, Gong J, Li L, Meng Y. Healing effects of a protein scaffold loaded with adipose-derived mesenchymal stem cells on radiation-induced vaginal injury in rats. J Int Med Res 2021; 48:300060520958826. [PMID: 33115306 PMCID: PMC7607296 DOI: 10.1177/0300060520958826] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Objectives Cervical cancer, the most common female cancer after breast cancer, is
typically treated using radiotherapy. However, pelvic radiotherapy can cause
irreversible damage to the vagina, seriously affecting patients’ quality of
life. In this study, protein scaffolds loaded with rat adipose-derived
mesenchymal stem cells (ADSCs) were implanted into irradiated tissue to
assess their healing potential. Methods We established a rat model of radiation-induced vaginal injury. Complexes
(consisting of protein scaffolds loaded with ADSCs) were implanted into
injury sites. Histological analysis were used to assess regeneration of the
vaginal epithelium. RNA sequencing was used to study the therapeutic
mechanism of the complexes. Results The complexes promoted vaginal epithelial cell regeneration, vaginal tissue
repair and improved vaginal stenosis and contracture. Compared with rats
transplanted with ADSCs, rats transplanted with complexes achieved better
therapeutic effects. Conclusions Protein scaffold-ADSC complexes had a beneficial therapeutic effect on
radiation-induced vaginal injury in rats and may serve as the basis of a
novel therapeutic approach for radiation dermatitis.
Collapse
Affiliation(s)
- Mingxia Ye
- Medical School of Chinese PLA, Department of Obstetrics and Gynecology The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Ling Yu
- Department of Obstetrics and Gynecology, The First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Yujia She
- Department of Obstetrics and Gynecology, The First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Shufang Wang
- Blood Transfusion Department, The First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Min Wang
- Medical School of Chinese PLA, Department of Obstetrics and Gynecology The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Qingdong Zhao
- Medical School of Chinese PLA, Department of Obstetrics and Gynecology The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Chenglei Gu
- The Eighth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Lihua Bian
- Department of Obstetrics and Gynecology, The First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Na Wen
- Medical School of Chinese PLA, Department of Obstetrics and Gynecology The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Jing Gong
- Anzhen Hospital Affiliated to Capital Medical University, Beijing, China
| | - Lian Li
- Department of Obstetrics and Gynecology, The First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Yuanguang Meng
- Department of Obstetrics and Gynecology, The First Medical Center of Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
16
|
Chen L, Liu G, Li W, Wu X. Synergistic effects of Indian hedgehog and sonic hedgehog on chondrogenesis during cartilage repair. J Mol Histol 2021; 52:407-418. [PMID: 33598817 DOI: 10.1007/s10735-021-09964-2] [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: 03/29/2020] [Accepted: 02/09/2021] [Indexed: 11/26/2022]
Abstract
Sonic hedgehog (Shh) and Indian hedgehog (Ihh) have been shown to control the induction of early cartilaginous differentiation. However, it is unclear whether Ihh and Shh exert synergistic effects on chondrogenesis during articular cartilage repair. Herein, we investigate the effects of chondrogenesis of bone-derived mesenchymal stem cells (BMSCs) following co-transfection with Shh and Ihh via adenoviral vectors in vitro and in vivo. A rotary cell culture system (RCCS) and Cytodex 3 microcarriers were used to create a stereoscopic dynamic environment for cell culture. In the RCCS environment, BMSCs co-transfected with Ihh and Shh displayed stronger chondrogenic differentiation and chondrogenesis than BMSCs transfected with Ihh or Shh alone, and exhibited higher expression levels of Sox 9, ACAN and collagen II, stronger toluidine blue and collagen II immunohistochemical staining. After transplanted into the osteochondral defect at 8 weeks, Ihh/Shh co-transfected BMSCs showed a significantly better cartilage repair than BMSCs transfected with Ihh or Shh alone. Ihh and Shh have synergistic effects on the induction of chondrogenic differentiation and chondrogenesis under a microgravity environment, and help to repair damaged cartilage and reverse subchondral defects during the early stages.
Collapse
Affiliation(s)
- Liyang Chen
- Department of Orthopaedics, Shanghai Tenth People's Hospital, Tongji University, Shanghai, 200072, China
- Shanghai Pudong Hospital, Fudan University, Shanghai, 201399, China
| | - Gejun Liu
- Department of Orthopaedics, Shanghai Tenth People's Hospital, Tongji University, Shanghai, 200072, China
| | - Wenjun Li
- Department of Orthopaedics, Shanghai Tenth People's Hospital, Tongji University, Shanghai, 200072, China
| | - Xing Wu
- Department of Orthopaedics, Shanghai Tenth People's Hospital, Tongji University, Shanghai, 200072, China.
| |
Collapse
|
17
|
Su Y, Liu Y, Ma C, Guan C, Ma X, Meng S. Mesenchymal stem cell-originated exosomal lncRNA HAND2-AS1 impairs rheumatoid arthritis fibroblast-like synoviocyte activation through miR-143-3p/TNFAIP3/NF-κB pathway. J Orthop Surg Res 2021; 16:116. [PMID: 33549125 PMCID: PMC7866436 DOI: 10.1186/s13018-021-02248-1] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 01/18/2021] [Indexed: 02/07/2023] Open
Abstract
Background Long non-coding RNA heart and neural crest derivatives expressed 2-antisense RNA 1 (HAND2-AS1) was found to be elevated in rheumatoid arthritis (RA) fibroblast-like synoviocytes (RA-FLSs). However, whether HAND2-AS1 functions as an exosomal lncRNA related to mesenchymal stem cells (MSCs) in RA progression is unknown. Methods The expression of HAND2-AS1, microRNA (miR)-143-3p, and tumor necrosis factor alpha-inducible protein 3 (TNFAIP3) was detected using quantitative real-time polymerase chain reaction and Western blot. Cell proliferation, apoptosis, migration, and invasion were detected using cell counting kit-8, flow cytometry, and wound healing and transwell assays. The levels of tumor necrosis factor-α (TNF-α) and interleukins (IL)-6 were analyzed using enzyme-linked immunosorbent assay. The level of phosphorylated-p65 was examined by Western blot. The binding interaction between miR-143-3p and HAND2-AS1 or TNFAIP3 was confirmed by the dual-luciferase reporter and RIP assays. Exosomes were isolated by ultracentrifugation and qualified by transmission electron microscopy (TEM), nanoparticle tracking analysis (NTA), and Western blot. Results HAND2-AS1 was lowly expressed in RA synovial tissues, and HAND2-AS1 re-expression suppressed the proliferation, motility, and inflammation and triggered the apoptosis in RA-FLSs via the inactivation of NF-κB pathway. Mechanistically, HAND2-AS1 directly sponged miR-143-3p and positively regulated TNFAIP3 expression, the target of miR-143-3p. Moreover, the effects of HAND2-AS1 on RA-FLSs were partially attenuated by miR-143-3p upregulation or TNFAIP3 knockdown. HAND2-AS1 could be packaged into hMSC-derived exosomes and absorbed by RA-FLSs, and human MSC-derived exosomal HAND2-AS1 also repressed above malignant biological behavior of RA-FLSs. Conclusion MSC-derived exosomes participated in the intercellular transfer of HAND2-AS1 and suppressed the activation of RA-FLSs via miR-143-3p/TNFAIP3/NF-κB pathway, which provided a novel insight into the pathogenesis and treatment of RA.
Collapse
Affiliation(s)
- Yuhua Su
- Department of Rheumatology and Immunology, Affiliated Hospital of Weifang Medical University, NO.2428 Yuhe Road, Kuiwen District, Weifang, 261000, Shandong, China
| | - Yajing Liu
- Department of Rheumatology and Immunology, Affiliated Hospital of Weifang Medical University, NO.2428 Yuhe Road, Kuiwen District, Weifang, 261000, Shandong, China
| | - Chao Ma
- Internal medicine, Yuncheng Hospital of traditional Chinese Medicine, Heze, 274700, Shandong, China
| | - Chunxiao Guan
- Department of Rheumatology and Immunology, Affiliated Hospital of Weifang Medical University, NO.2428 Yuhe Road, Kuiwen District, Weifang, 261000, Shandong, China
| | - Xiufen Ma
- Department of Rheumatology and Immunology, Affiliated Hospital of Weifang Medical University, NO.2428 Yuhe Road, Kuiwen District, Weifang, 261000, Shandong, China
| | - Shan Meng
- Department of Rheumatology and Immunology, Affiliated Hospital of Weifang Medical University, NO.2428 Yuhe Road, Kuiwen District, Weifang, 261000, Shandong, China.
| |
Collapse
|
18
|
Davis S, Roldo M, Blunn G, Tozzi G, Roncada T. Influence of the Mechanical Environment on the Regeneration of Osteochondral Defects. Front Bioeng Biotechnol 2021; 9:603408. [PMID: 33585430 PMCID: PMC7873466 DOI: 10.3389/fbioe.2021.603408] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2020] [Accepted: 01/04/2021] [Indexed: 12/20/2022] Open
Abstract
Articular cartilage is a highly specialised connective tissue of diarthrodial joints which provides a smooth, lubricated surface for joint articulation and plays a crucial role in the transmission of loads. In vivo cartilage is subjected to mechanical stimuli that are essential for cartilage development and the maintenance of a chondrocytic phenotype. Cartilage damage caused by traumatic injuries, ageing, or degradative diseases leads to impaired loading resistance and progressive degeneration of both the articular cartilage and the underlying subchondral bone. Since the tissue has limited self-repairing capacity due its avascular nature, restoration of its mechanical properties is still a major challenge. Tissue engineering techniques have the potential to heal osteochondral defects using a combination of stem cells, growth factors, and biomaterials that could produce a biomechanically functional tissue, representative of native hyaline cartilage. However, current clinical approaches fail to repair full-thickness defects that include the underlying subchondral bone. Moreover, when tested in vivo, current tissue-engineered grafts show limited capacity to regenerate the damaged tissue due to poor integration with host cartilage and the failure to retain structural integrity after insertion, resulting in reduced mechanical function. The aim of this review is to examine the optimal characteristics of osteochondral scaffolds. Additionally, an overview on the latest biomaterials potentially able to replicate the natural mechanical environment of articular cartilage and their role in maintaining mechanical cues to drive chondrogenesis will be detailed, as well as the overall mechanical performance of grafts engineered using different technologies.
Collapse
Affiliation(s)
- Sarah Davis
- School of Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth, United Kingdom
| | - Marta Roldo
- School of Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth, United Kingdom
| | - Gordon Blunn
- School of Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth, United Kingdom
| | - Gianluca Tozzi
- Zeiss Global Centre, School of Mechanical and Design Engineering, University of Portsmouth, Portsmouth, United Kingdom
| | - Tosca Roncada
- School of Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth, United Kingdom
| |
Collapse
|
19
|
Spampinato M, Giallongo C, Romano A, Longhitano L, La Spina E, Avola R, Scandura G, Dulcamare I, Bramanti V, Di Rosa M, Vicario N, Parenti R, Li Volti G, Tibullo D, Palumbo GA. Focus on Osteosclerotic Progression in Primary Myelofibrosis. Biomolecules 2021; 11:biom11010122. [PMID: 33477816 PMCID: PMC7832894 DOI: 10.3390/biom11010122] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 01/09/2021] [Accepted: 01/16/2021] [Indexed: 12/22/2022] Open
Abstract
Primary myelofibrosis (PMF) is a myeloproliferative neoplasm characterized by hematopoietic stem-cell-derived clonal proliferation, leading to bone marrow (BM) fibrosis. Hematopoiesis alterations are closely associated with modifications of the BM microenvironment, characterized by defective interactions between vascular and endosteal niches. As such, neoangiogenesis, megakaryocytes hyperplasia and extensive bone marrow fibrosis, followed by osteosclerosis and bone damage, are the most relevant consequences of PMF. Moreover, bone tissue deposition, together with progressive fibrosis, represents crucial mechanisms of disabilities in patients. Although the underlying mechanisms of bone damage observed in PMF are still unclear, the involvement of cytokines, growth factors and bone marrow microenvironment resident cells have been linked to disease progression. Herein, we focused on the role of megakaryocytes and their alterations, associated with cytokines and chemokines release, in modulating functions of most of the bone marrow cell populations and in creating a complex network where impaired signaling strongly contributes to progression and disabilities.
Collapse
Affiliation(s)
- Mariarita Spampinato
- Section of Biochemistry, Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (M.S.); (L.L.); (R.A.); (D.T.)
| | - Cesarina Giallongo
- Department of Medical and Surgical Sciences and Advanced Technologies “G.F. Ingrassia”, University of Catania, 95123 Catania, Italy;
| | - Alessandra Romano
- Department of General Surgery and Medical-Surgical Specialties, Division of Hematology, A.O.U. “Policlinico-Vittorio Emanuele”, University of Catania, 95123 Catania, Italy; (A.R.); (E.L.S.); (G.S.); (I.D.)
| | - Lucia Longhitano
- Section of Biochemistry, Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (M.S.); (L.L.); (R.A.); (D.T.)
| | - Enrico La Spina
- Department of General Surgery and Medical-Surgical Specialties, Division of Hematology, A.O.U. “Policlinico-Vittorio Emanuele”, University of Catania, 95123 Catania, Italy; (A.R.); (E.L.S.); (G.S.); (I.D.)
| | - Roberto Avola
- Section of Biochemistry, Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (M.S.); (L.L.); (R.A.); (D.T.)
| | - Grazia Scandura
- Department of General Surgery and Medical-Surgical Specialties, Division of Hematology, A.O.U. “Policlinico-Vittorio Emanuele”, University of Catania, 95123 Catania, Italy; (A.R.); (E.L.S.); (G.S.); (I.D.)
| | - Ilaria Dulcamare
- Department of General Surgery and Medical-Surgical Specialties, Division of Hematology, A.O.U. “Policlinico-Vittorio Emanuele”, University of Catania, 95123 Catania, Italy; (A.R.); (E.L.S.); (G.S.); (I.D.)
| | - Vincenzo Bramanti
- Division of Clinical Pathology, “Giovanni Paolo II” Hospital–A.S.P. Ragusa, 97100 Ragusa, Italy;
| | - Michelino Di Rosa
- Section of Human Anatomy and Histology, Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy;
| | - Nunzio Vicario
- Section of Physiology, Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (N.V.); (R.P.)
| | - Rosalba Parenti
- Section of Physiology, Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (N.V.); (R.P.)
| | - Giovanni Li Volti
- Section of Biochemistry, Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (M.S.); (L.L.); (R.A.); (D.T.)
- Correspondence: (G.L.V.); (G.A.P.)
| | - Daniele Tibullo
- Section of Biochemistry, Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (M.S.); (L.L.); (R.A.); (D.T.)
| | - Giuseppe A. Palumbo
- Department of Medical and Surgical Sciences and Advanced Technologies “G.F. Ingrassia”, University of Catania, 95123 Catania, Italy;
- Correspondence: (G.L.V.); (G.A.P.)
| |
Collapse
|
20
|
Spampinato M, Giallongo C, Romano A, Longhitano L, La Spina E, Avola R, Scandura G, Dulcamare I, Bramanti V, Di Rosa M, Vicario N, Parenti R, Li Volti G, Tibullo D, Palumbo GA. Focus on Osteosclerotic Progression in Primary Myelofibrosis. Biomolecules 2021. [PMID: 33477816 DOI: 10.3390/biom11010122.pmid:33477816;pmcid:pmc7832894] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/09/2023] Open
Abstract
Primary myelofibrosis (PMF) is a myeloproliferative neoplasm characterized by hematopoietic stem-cell-derived clonal proliferation, leading to bone marrow (BM) fibrosis. Hematopoiesis alterations are closely associated with modifications of the BM microenvironment, characterized by defective interactions between vascular and endosteal niches. As such, neoangiogenesis, megakaryocytes hyperplasia and extensive bone marrow fibrosis, followed by osteosclerosis and bone damage, are the most relevant consequences of PMF. Moreover, bone tissue deposition, together with progressive fibrosis, represents crucial mechanisms of disabilities in patients. Although the underlying mechanisms of bone damage observed in PMF are still unclear, the involvement of cytokines, growth factors and bone marrow microenvironment resident cells have been linked to disease progression. Herein, we focused on the role of megakaryocytes and their alterations, associated with cytokines and chemokines release, in modulating functions of most of the bone marrow cell populations and in creating a complex network where impaired signaling strongly contributes to progression and disabilities.
Collapse
Affiliation(s)
- Mariarita Spampinato
- Section of Biochemistry, Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy
| | - Cesarina Giallongo
- Department of Medical and Surgical Sciences and Advanced Technologies "G.F. Ingrassia", University of Catania, 95123 Catania, Italy
| | - Alessandra Romano
- Department of General Surgery and Medical-Surgical Specialties, Division of Hematology, A.O.U. "Policlinico-Vittorio Emanuele", University of Catania, 95123 Catania, Italy
| | - Lucia Longhitano
- Section of Biochemistry, Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy
| | - Enrico La Spina
- Department of General Surgery and Medical-Surgical Specialties, Division of Hematology, A.O.U. "Policlinico-Vittorio Emanuele", University of Catania, 95123 Catania, Italy
| | - Roberto Avola
- Section of Biochemistry, Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy
| | - Grazia Scandura
- Department of General Surgery and Medical-Surgical Specialties, Division of Hematology, A.O.U. "Policlinico-Vittorio Emanuele", University of Catania, 95123 Catania, Italy
| | - Ilaria Dulcamare
- Department of General Surgery and Medical-Surgical Specialties, Division of Hematology, A.O.U. "Policlinico-Vittorio Emanuele", University of Catania, 95123 Catania, Italy
| | - Vincenzo Bramanti
- Division of Clinical Pathology, "Giovanni Paolo II" Hospital-A.S.P. Ragusa, 97100 Ragusa, Italy
| | - Michelino Di Rosa
- Section of Human Anatomy and Histology, Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy
| | - Nunzio Vicario
- Section of Physiology, Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy
| | - Rosalba Parenti
- Section of Physiology, Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy
| | - Giovanni Li Volti
- Section of Biochemistry, Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy
| | - Daniele Tibullo
- Section of Biochemistry, Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy
| | - Giuseppe A Palumbo
- Department of Medical and Surgical Sciences and Advanced Technologies "G.F. Ingrassia", University of Catania, 95123 Catania, Italy
| |
Collapse
|
21
|
Sohrabi M, Eftekhari Yekta B, Rezaie H, Naimi-Jamal MR, Kumar A, Cochis A, Miola M, Rimondini L. Enhancing Mechanical Properties and Biological Performances of Injectable Bioactive Glass by Gelatin and Chitosan for Bone Small Defect Repair. Biomedicines 2020; 8:biomedicines8120616. [PMID: 33334044 PMCID: PMC7765522 DOI: 10.3390/biomedicines8120616] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 12/07/2020] [Accepted: 12/13/2020] [Indexed: 12/19/2022] Open
Abstract
Bioactive glass (BG) represents a promising biomaterial for bone healing; here injectable BG pastes biological properties were improved by the addition of gelatin or chitosan, as well as mechanical resistance was enhanced by adding 10 or 20 wt% 3-Glycidyloxypropyl trimethoxysilane (GPTMS) cross-linker. Composite pastes exhibited bioactivity as apatite formation was observed by Scanning Electron Microscopy (SEM) and X-Ray Diffraction (XRD) after 14 days immersion in simulated body fluid (SBF); moreover, polymers did not enhance degradability as weight loss was >10% after 30 days in physiological conditions. BG-gelatin-20 wt% GPTMS composites demonstrated the highest compressive strength (4.8 ± 0.5 MPa) in comparison with the bulk control paste made of 100% BG in water (1.9 ± 0.1 MPa). Cytocompatibility was demonstrated towards human mesenchymal stem cells (hMSC), osteoblasts progenitors, and endothelial cells. The presence of 20 wt% GPTMS conferred antibacterial properties thus inhibiting the joint pathogens Staphylococcus aureus and Staphylococcus epidermidis infection. Finally, hMSC osteogenesis was successfully supported in a 3D model as demonstrated by alkaline phosphatase release and osteogenic genes expression.
Collapse
Affiliation(s)
- Mehri Sohrabi
- School of Metallurgy and Materials Engineering, Iran University of Science and Technology, Tehran 1684613114, Iran; (M.S.); (H.R.)
| | - Bijan Eftekhari Yekta
- School of Metallurgy and Materials Engineering, Iran University of Science and Technology, Tehran 1684613114, Iran; (M.S.); (H.R.)
- Correspondence: (B.E.Y.); (L.R.)
| | - Hamidreza Rezaie
- School of Metallurgy and Materials Engineering, Iran University of Science and Technology, Tehran 1684613114, Iran; (M.S.); (H.R.)
| | - Mohammad Reza Naimi-Jamal
- Department of Chemistry, Research Laboratory of Green Organic Synthesis and Polymers, Iran University of Science and Technology, Tehran 1684613114, Iran;
| | - Ajay Kumar
- Department of Health Sciences, Center for Translational Research on Autoimmune and Allergic Diseases–CAAD, University of Piemonte Orientale UPO, 28100 Novara, Italy; (A.K.); (A.C.)
| | - Andrea Cochis
- Department of Health Sciences, Center for Translational Research on Autoimmune and Allergic Diseases–CAAD, University of Piemonte Orientale UPO, 28100 Novara, Italy; (A.K.); (A.C.)
| | - Marta Miola
- Institute of Materials Engineering and Physics, Department of Applied Science and Technology, Politecnico di Torino, 10129 Turin, Italy;
| | - Lia Rimondini
- Department of Health Sciences, Center for Translational Research on Autoimmune and Allergic Diseases–CAAD, University of Piemonte Orientale UPO, 28100 Novara, Italy; (A.K.); (A.C.)
- Correspondence: (B.E.Y.); (L.R.)
| |
Collapse
|
22
|
Gallo N, Natali ML, Sannino A, Salvatore L. An Overview of the Use of Equine Collagen as Emerging Material for Biomedical Applications. J Funct Biomater 2020; 11:79. [PMID: 33139660 PMCID: PMC7712325 DOI: 10.3390/jfb11040079] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 10/24/2020] [Accepted: 10/29/2020] [Indexed: 12/19/2022] Open
Abstract
Type I collagen has always aroused great interest in the field of life-science and bioengineering, thanks to its favorable structural properties and bioactivity. For this reason, in the last five decades it has been widely studied and employed as biomaterial for the manufacture of implantable medical devices. Commonly used sources of collagen are represented by bovine and swine but their applications are limited because of the zoonosis transmission risks, the immune response and the religious constrains. Thus, type-I collagen isolated from horse tendon has recently gained increasing interest as an attractive alternative, so that, although bovine and porcine derived collagens still remain the most common ones, more and more companies started to bring to market a various range of equine collagen-based products. In this context, this work aims to overview the properties of equine collagen making it particularly appealing in medicine, cosmetics and pharmaceuticals, as well as its main biomedical applications and the currently approved equine collagen-based medical devices, focusing on experimental studies and clinical trials of the last 15 years. To the best of our knowledge, this is the first review focusing on the use of equine collagen, as well as on equine collagen-based marketed products for healthcare.
Collapse
Affiliation(s)
- Nunzia Gallo
- Department of Engineering for Innovation, University of Salento, Via Monteroni, 73100 Lecce, Italy; (M.L.N.); (A.S.); (L.S.)
| | | | | | | |
Collapse
|
23
|
An insight on type I collagen from horse tendon for the manufacture of implantable devices. Int J Biol Macromol 2020; 154:291-306. [DOI: 10.1016/j.ijbiomac.2020.03.082] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Accepted: 03/11/2020] [Indexed: 12/14/2022]
|
24
|
Meng Q, Qiu B. Exosomal MicroRNA-320a Derived From Mesenchymal Stem Cells Regulates Rheumatoid Arthritis Fibroblast-Like Synoviocyte Activation by Suppressing CXCL9 Expression. Front Physiol 2020; 11:441. [PMID: 32528301 PMCID: PMC7264418 DOI: 10.3389/fphys.2020.00441] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Accepted: 04/08/2020] [Indexed: 12/20/2022] Open
Abstract
Rheumatoid arthritis (RA), a chronic systemic inflammatory disease, is a primary cause of disability worldwide. The involvement of fibroblast-like synoviocytes (FLSs) in the regulation of the pathogenesis of RA has been highlighted. Mesenchymal stem cells (MSCs) are important candidates for cell-based treatment in many inflammatory autoimmune diseases. Herein, we identify whether MSC-derived exosomes loaded with microRNA-320a (miR-320a) regulate RA-FLSs. Synovial tissues from 22 patients with RA and 9 patients with osteoarthritis were collected. RA-FLSs were obtained from patients with RA, and their functions were evaluated by determining levels of interleukin-1β (IL-1β), IL-6, and IL-8 and by transwell migration and invasion assays. Dual luciferase reporter gene assays were employed to identify interaction between miR-320a and CXC chemokine ligand 9 (CXCL9). A co-culture system of MSC-derived exosomes and RA-FLSs were performed. The collagen-induced arthritis (CIA) mouse models with arthritis and bone damage were developed. Our results revealed the existence of reciprocal expression of miR-320a and CXCL9 in the synovial tissues obtained from patients with RA. CXCL9 knockdown or miR-320a upregulation suppressed the activation, migration, and invasion of RA-FLSs. CXCL9 was confirmed to be a target of miR-320a, and CXCL9 overexpression restored RA-FLS function in the presence of miR-320a. MSC-derived exosomes containing miR-320a mimic significantly suppressed RA-FLS activation, migration, and invasion in vitro and attenuated arthritis and bone damage in mice with CIA in vivo. Our study uncovers that MSC-derived exosomes participate in the intercellular transfer of miR-320a and subsequently inhibit the progression of RA. These results provide a novel potential therapeutic approach for RA treatment by increasing miR-320a in exosomes.
Collapse
Affiliation(s)
- Qing Meng
- Department of Orthopedics, Guizhou Orthopedics Hospital, Guiyang, China
| | - Bing Qiu
- Department of Orthopedics, Guizhou Orthopedics Hospital, Guiyang, China
| |
Collapse
|
25
|
Szychlinska MA, Calabrese G, Ravalli S, Dolcimascolo A, Castrogiovanni P, Fabbi C, Puglisi C, Lauretta G, Di Rosa M, Castorina A, Parenti R, Musumeci G. Evaluation of a Cell-Free Collagen Type I-Based Scaffold for Articular Cartilage Regeneration in an Orthotopic Rat Model. MATERIALS 2020; 13:ma13102369. [PMID: 32455683 PMCID: PMC7287598 DOI: 10.3390/ma13102369] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 05/16/2020] [Accepted: 05/19/2020] [Indexed: 12/23/2022]
Abstract
The management of chondral defects represents a big challenge because of the limited self-healing capacity of cartilage. Many approaches in this field obtained partial satisfactory results. Cartilage tissue engineering, combining innovative scaffolds and stem cells from different sources, emerges as a promising strategy for cartilage regeneration. The aim of this study was to evaluate the capability of a cell-free collagen I-based scaffold to promote cartilaginous repair after orthotopic implantation in vivo. Articular cartilage lesions (ACL) were created at the femoropatellar groove in rat knees and cell free collagen I-based scaffolds (S) were then implanted into right knee defect for the ACL-S group. No scaffold was implanted for the ACL group. At 4-, 8- and 16-weeks post-transplantation, degrees of cartilage repair were evaluated by morphological, histochemical and gene expression analyses. Histological analysis shows the formation of fibrous tissue, at 4-weeks replaced by a tissue resembling the calcified one at 16-weeks in the ACL group. In the ACL-S group, progressive replacement of the scaffold with the newly formed cartilage-like tissue is shown, as confirmed by Alcian Blue staining. Immunohistochemical and quantitative real-time PCR (qRT-PCR) analyses display the expression of typical cartilage markers, such as collagen type I and II (ColI and ColII), Aggrecan and Sox9. The results of this study display that the collagen I-based scaffold is highly biocompatible and able to recruit host cells from the surrounding joint tissues to promote cartilaginous repair of articular defects, suggesting its use as a potential approach for cartilage tissue regeneration.
Collapse
Affiliation(s)
- Marta Anna Szychlinska
- Department of Biomedical and Biotechnological Sciences, Anatomy, Histology and Movement Sciences Section, School of Medicine, University of Catania, 95123 Catania, Italy; (M.A.S.); (S.R.); (P.C.); (G.L.); (M.D.R.)
| | - Giovanna Calabrese
- Department of Biomedical and Biotechnological Sciences, Physiology Section, School of Medicine, University of Catania, 95123 Catania, Italy; (G.C.); (A.D.); (R.P.)
| | - Silvia Ravalli
- Department of Biomedical and Biotechnological Sciences, Anatomy, Histology and Movement Sciences Section, School of Medicine, University of Catania, 95123 Catania, Italy; (M.A.S.); (S.R.); (P.C.); (G.L.); (M.D.R.)
| | - Anna Dolcimascolo
- Department of Biomedical and Biotechnological Sciences, Physiology Section, School of Medicine, University of Catania, 95123 Catania, Italy; (G.C.); (A.D.); (R.P.)
| | - Paola Castrogiovanni
- Department of Biomedical and Biotechnological Sciences, Anatomy, Histology and Movement Sciences Section, School of Medicine, University of Catania, 95123 Catania, Italy; (M.A.S.); (S.R.); (P.C.); (G.L.); (M.D.R.)
| | | | - Caterina Puglisi
- Istituto Oncologico del Mediterraneo (IOM), 95029 Viagrande, 95123 Catania, Italy;
| | - Giovanni Lauretta
- Department of Biomedical and Biotechnological Sciences, Anatomy, Histology and Movement Sciences Section, School of Medicine, University of Catania, 95123 Catania, Italy; (M.A.S.); (S.R.); (P.C.); (G.L.); (M.D.R.)
| | - Michelino Di Rosa
- Department of Biomedical and Biotechnological Sciences, Anatomy, Histology and Movement Sciences Section, School of Medicine, University of Catania, 95123 Catania, Italy; (M.A.S.); (S.R.); (P.C.); (G.L.); (M.D.R.)
| | - Alessandro Castorina
- School of Life Science, Faculty of Science, University of Technology Sydney, Sydney, NSW 123, Australia;
- Discipline of Anatomy & Histology, School of Medical Sciences, The University of Sydney, Sydney, NSW 123, Australia
| | - Rosalba Parenti
- Department of Biomedical and Biotechnological Sciences, Physiology Section, School of Medicine, University of Catania, 95123 Catania, Italy; (G.C.); (A.D.); (R.P.)
| | - Giuseppe Musumeci
- Department of Biomedical and Biotechnological Sciences, Anatomy, Histology and Movement Sciences Section, School of Medicine, University of Catania, 95123 Catania, Italy; (M.A.S.); (S.R.); (P.C.); (G.L.); (M.D.R.)
- Research Center on Motor Activities (CRAM), University of Catania, 95123 Catania, Italy
- Department of Biology, Sbarro Institute for Cancer Research and Molecular Medicine, College of Science and Technology, Temple University, Philadelphia, PA 19122, USA
- Correspondence: ; Tel.: +095-378-2036
| |
Collapse
|
26
|
Kronemberger GS, Matsui RAM, Miranda GDASDCE, Granjeiro JM, Baptista LS. Cartilage and bone tissue engineering using adipose stromal/stem cells spheroids as building blocks. World J Stem Cells 2020; 12:110-122. [PMID: 32184936 PMCID: PMC7062040 DOI: 10.4252/wjsc.v12.i2.110] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 10/19/2019] [Accepted: 01/15/2020] [Indexed: 02/06/2023] Open
Abstract
Scaffold-free techniques in the developmental tissue engineering area are designed to mimic in vivo embryonic processes with the aim of biofabricating, in vitro, tissues with more authentic properties. Cell clusters called spheroids are the basis for scaffold-free tissue engineering. In this review, we explore the use of spheroids from adult mesenchymal stem/stromal cells as a model in the developmental engineering area in order to mimic the developmental stages of cartilage and bone tissues. Spheroids from adult mesenchymal stromal/stem cells lineages recapitulate crucial events in bone and cartilage formation during embryogenesis, and are capable of spontaneously fusing to other spheroids, making them ideal building blocks for bone and cartilage tissue engineering. Here, we discuss data from ours and other labs on the use of adipose stromal/stem cell spheroids in chondrogenesis and osteogenesis in vitro. Overall, recent studies support the notion that spheroids are ideal "building blocks" for tissue engineering by “bottom-up” approaches, which are based on tissue assembly by advanced techniques such as three-dimensional bioprinting. Further studies on the cellular and molecular mechanisms that orchestrate spheroid fusion are now crucial to support continued development of bottom-up tissue engineering approaches such as three-dimensional bioprinting.
Collapse
Affiliation(s)
- Gabriela S Kronemberger
- Laboratory of Tissue Bioengineering, Directory of Metrology Applied to Life Sciences, National Institute of Metrology, Quality and Technology (INMETRO), Duque de Caxias, RJ 25250-020, Brazil
- Post-graduate Program in Translational Biomedicine (Biotrans), Unigranrio, Campus I, Duque de Caxias, RJ 25250-020, Brazil
| | - Renata Akemi Morais Matsui
- Laboratory of Tissue Bioengineering, Directory of Metrology Applied to Life Sciences, National Institute of Metrology, Quality and Technology (INMETRO), Duque de Caxias, RJ 25250-020, Brazil
- Post-graduate Program in Biotechnology, National Institute of Metrology, Quality and Technology (INMETRO), Duque de Caxias, RJ 25250-020, Brazil
| | - Guilherme de Almeida Santos de Castro e Miranda
- Laboratory of Tissue Bioengineering, Directory of Metrology Applied to Life Sciences, National Institute of Metrology, Quality and Technology (INMETRO), Duque de Caxias, RJ 25250-020, Brazil
- Federal University of Rio de Janeiro (UFRJ), Campus Duque de Caxias, Duque de Caxias, RJ 25250-020, Brazil
| | - José Mauro Granjeiro
- Laboratory of Tissue Bioengineering, Directory of Metrology Applied to Life Sciences, National Institute of Metrology, Quality and Technology (INMETRO), Duque de Caxias, RJ 25250-020, Brazil
- Post-graduate Program in Biotechnology, National Institute of Metrology, Quality and Technology (INMETRO), Duque de Caxias, RJ 25250-020, Brazil
- Laboratory of Clinical Research in Odontology, Fluminense Federal University (UFF), Niterói 25255-030 Brazil
| | - Leandra Santos Baptista
- Laboratory of Tissue Bioengineering, Directory of Metrology Applied to Life Sciences, National Institute of Metrology, Quality and Technology (INMETRO), Duque de Caxias, RJ 25250-020, Brazil
- Post-graduate Program in Translational Biomedicine (Biotrans), Unigranrio, Campus I, Duque de Caxias, RJ 25250-020, Brazil
- Post-graduate Program in Biotechnology, National Institute of Metrology, Quality and Technology (INMETRO), Duque de Caxias, RJ 25250-020, Brazil
- Multidisciplinary Center for Biological Research (Numpex-Bio), Federal University of Rio de Janeiro (UFRJ) Campus Duque de Caxias, Duque de Caxias, RJ 25245-390, Brazil
| |
Collapse
|
27
|
Bao W, Li M, Yang Y, Wan Y, Wang X, Bi N, Li C. Advancements and Frontiers in the High Performance of Natural Hydrogels for Cartilage Tissue Engineering. Front Chem 2020; 8:53. [PMID: 32117879 PMCID: PMC7028759 DOI: 10.3389/fchem.2020.00053] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2019] [Accepted: 01/17/2020] [Indexed: 12/14/2022] Open
Abstract
Cartilage injury originating from trauma or osteoarthritis is a common joint disease that can bring about an increasing social and economic burden in modern society. On account of its avascular, neural, and lymphatic characteristics, the poor migration ability of chondrocytes, and a low number of progenitor cells, the self-healing ability of cartilage defects has been significantly limited. Natural hydrogels, occurring abundantly with characteristics such as high water absorption, biodegradation, adjustable porosity, and biocompatibility like that of the natural extracellular matrix (ECM), have been developed into one of the most suitable scaffold biomaterials for the regeneration of cartilage in material science and tissue engineering. Notably, natural hydrogels derived from sources such as animal or human cadaver tissues possess the bionic mechanical behaviors of physiological cartilage that are required for usage as articular cartilage substitutes, by which the enhanced chondrogenic phenotype ability may be achieved by facilely embedding living cells, controlling degradation profiles, and releasing stimulatory growth factors. Hence, we summarize an overview of strategies and developments of the various kinds and functions of natural hydrogels for cartilage tissue engineering in this review. The main concepts and recent essential research found that great challenges like vascularity, clinically relevant size, and mechanical performances were still difficult to overcome because the current limitations of technologies need to be severely addressed in practical settings, particularly in unpredictable preclinical trials and during future forays into cartilage regeneration using natural hydrogel scaffolds with high mechanical properties. Therefore, the grand aim of this current review is to underpin the importance of preparation, modification, and application for the high performance of natural hydrogels for cartilage tissue engineering, which has been achieved by presenting a promising avenue in various fields and postulating real-world respective potentials.
Collapse
Affiliation(s)
- Wuren Bao
- School of Nursing, Inner Mongolia University for Nationalities, Tongliao, China
| | - Menglu Li
- Beijing National Laboratory for Molecular Sciences, State Key Laboratory of Polymer Physics & Chemistry, Institute of Chemistry, Chinese Academy of Sciences, Beijing, China
| | - Yanyu Yang
- Beijing National Laboratory for Molecular Sciences, State Key Laboratory of Polymer Physics & Chemistry, Institute of Chemistry, Chinese Academy of Sciences, Beijing, China
- College of Science and Engineering, Zhengzhou University, Zhengzhou, China
| | - Yi Wan
- Orthopaedic Department, The 8th Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Xing Wang
- Beijing National Laboratory for Molecular Sciences, State Key Laboratory of Polymer Physics & Chemistry, Institute of Chemistry, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Na Bi
- Orthopaedic Department, The 8th Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Chunlin Li
- Orthopaedic Department, The 8th Medical Center of Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
28
|
Elshishiny F, Mamdouh W. Fabrication of Nanofibrous/Xerogel Layer-by-Layer Biocomposite Scaffolds for Skin Tissue Regeneration: In Vitro Study. ACS OMEGA 2020; 5:2133-2147. [PMID: 32064374 PMCID: PMC7016933 DOI: 10.1021/acsomega.9b02832] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Accepted: 01/20/2020] [Indexed: 06/10/2023]
Abstract
Skin burn wounds are a crucial issue that could reduce life quality. Although numerous effective skin products have invaded the biomedical market, most of them still demonstrate some limitations regarding their porosity, swelling and degradation behaviors, antibacterial properties, and cytotoxicity. Thus, the aim of this study is to fabricate novel trilayered asymmetric porous scaffolds that can mimic the natural skin layers. In particular, the fabricated scaffold constitutes an upper electrospun chitosan-poly(vinyl alcohol) layer and a lower xerogel layer, which is made of effective skin extracellular matrix components. Both layers are fixed together using fibrin glue as a middle layer. The results of this study revealed promising scaffold swelling capability suitable for absorbing wound exudates, followed by a constant degradable weight over time, which is appropriate for a burn wound environment. Scanning electron microscopy images revealed an average pore diameter in the range of 138.39-170.18 nm for the cross-linked electrospun mats and an average pore size of 2.29-30.62 μm for the fabricated xerogel layers. This further provided an optimum environment for fibroblast migration and proliferation. The electrospun nanofibrous layer was examined for its antibacterial properties and showed expressive complete bacterial inhibition against Gram-positive (Staphylococcus aureus) and Gram-negative (Escherichia coli) bacterial strains (log reduction = 3 and 2.70, respectively). Next, mouse embryonic fibroblast cytotoxicity and migration rate were investigated against the developed asymmetrical composite to assess its biocompatibility. Tissue culture experiments demonstrated significant cell proliferation and migration in the presence of the constructed scaffold (P < 0.0001). A complete wound closure was observed in vitro in the presence of the three scaffold asymmetrical layers against the mouse embryonic fibroblast. The results of this study proved superior biological characteristics of the innovative asymmetrical composite that could further replace the burned or damaged skin layers with promising potential for clinical applications.
Collapse
Affiliation(s)
| | - Wael Mamdouh
- E-mail: . Tel: +202
2615 2555. Fax: +202 2797 4951
| |
Collapse
|
29
|
Cycloastragenol as an Exogenous Enhancer of Chondrogenic Differentiation of Human Adipose-Derived Mesenchymal Stem Cells. A Morphological Study. Cells 2020; 9:cells9020347. [PMID: 32028592 PMCID: PMC7072395 DOI: 10.3390/cells9020347] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 01/27/2020] [Accepted: 02/01/2020] [Indexed: 12/24/2022] Open
Abstract
Stem cell therapy and tissue engineering represent a promising approach for cartilage regeneration. However, they present limits in terms of mechanical properties and premature de-differentiation of engineered cartilage. Cycloastragenol (CAG), a triterpenoid saponin compound and a hydrolysis product of the main ingredient in Astragalus membranaceous, has been explored for cartilage regeneration. The aim of this study was to investigate CAG’s ability to promote cell proliferation, maintain cells in their stable active phenotype, and support the production of cartilaginous extracellular matrix (ECM) in human adipose-derived mesenchymal stem cells (hAMSCs) in up to 28 days of three-dimensional (3D) chondrogenic culture. The hAMSC pellets were cultured in chondrogenic medium (CM) and in CM supplemented with CAG (CAG–CM) for 7, 14, 21, and 28 days. At each time-point, the pellets were harvested for histological (hematoxylin and eosin (H&E)), histochemical (Alcian-Blue) and immunohistochemical analysis (Type I, II, and X collagen, aggrecan, SOX9, lubricin). After excluding CAG’s cytotoxicity (MTT Assay), improved cell condensation, higher glycosaminoglycans (sGAG) content, and increased cell proliferation have been detected in CAG–CM pellets until 28 days of culture. Overall, CAG improved the chondrogenic differentiation of hAMSCs, maintaining stable the active chondrocyte phenotype in up to 28 days of 3D in vitro chondrogenic culture. It is proposed that CAG might have a beneficial impact on cartilage regeneration approaches.
Collapse
|
30
|
Iqbal B, Muhammad N, Rahim A, Iqbal F, Sharif F, Safi SZ, Khan AS, Gonfa G, Uroos M, Rehman IU. Development of collagen/PVA composites patches for osteochondral defects using a green processing of ionic liquid. INT J POLYM MATER PO 2019; 68:590-596. [DOI: 10.1080/00914037.2018.1474358] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2017] [Accepted: 05/05/2018] [Indexed: 10/14/2022]
Affiliation(s)
- Bushra Iqbal
- Interdisciplinary Research Centre in Biomedical Materials (IRCBM), COMSATS Institute of Information Technology, Lahore, Pakistan
| | - Nawshad Muhammad
- Interdisciplinary Research Centre in Biomedical Materials (IRCBM), COMSATS Institute of Information Technology, Lahore, Pakistan
| | - Abdur Rahim
- Interdisciplinary Research Centre in Biomedical Materials (IRCBM), COMSATS Institute of Information Technology, Lahore, Pakistan
| | - Farasit Iqbal
- Interdisciplinary Research Centre in Biomedical Materials (IRCBM), COMSATS Institute of Information Technology, Lahore, Pakistan
| | - Faiza Sharif
- Interdisciplinary Research Centre in Biomedical Materials (IRCBM), COMSATS Institute of Information Technology, Lahore, Pakistan
| | - Sher Zaman Safi
- Interdisciplinary Research Centre in Biomedical Materials (IRCBM), COMSATS Institute of Information Technology, Lahore, Pakistan
| | - Amir Sada Khan
- Department of Chemical Engineering, Center for Research in Ionic Liquids, Universiti Teknologi PETRONAS (UTP), Tronoh, Perak, Malaysia
| | - Girma Gonfa
- College of Biological and Chemical Engineering Addis Ababa Science and Technology University, Addis Ababa, Ethiopia
| | - Maliha Uroos
- Department of Chemistry, University of the Punjab, Lahore, Pakistan
| | - Ihtesham Ur Rehman
- Department of Materials Science & Engineering, Kroto Research Institute, University of Sheffield, Sheffield, United Kingdom
| |
Collapse
|
31
|
Yamazaki T, Kishimoto T, Leszczyński P, Sadakane K, Kenmotsu T, Watanabe H, Kazama T, Matsumoto T, Yoshikawa K, Taniguchi H. Construction of 3D Cellular Composites with Stem Cells Derived from Adipose Tissue and Endothelial Cells by Use of Optical Tweezers in a Natural Polymer Solution. MATERIALS 2019; 12:ma12111759. [PMID: 31151204 PMCID: PMC6601048 DOI: 10.3390/ma12111759] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 05/17/2019] [Accepted: 05/22/2019] [Indexed: 02/06/2023]
Abstract
To better understand the regulation and function of cellular interactions, three-dimensional (3D) assemblies of single cells and subsequent functional analysis are gaining popularity in many research fields. While we have developed strategies to build stable cellular structures using optical tweezers in a minimally invasive state, methods for manipulating a wide range of cell types have yet to be established. To mimic organ-like structures, the construction of 3D cellular assemblies with variety of cell types is essential. Our recent studies have shown that the presence of nonspecific soluble polymers in aqueous solution is the key to creating stable 3D cellular assemblies efficiently. The present study further expands on the construction of 3D single cell assemblies using two different cell types. We have successfully generated 3D cellular assemblies, using GFP-labeled adipose tissue-derived stem cells and endothelial cells by using optical tweezers. Our findings will support the development of future applications to further characterize cellular interactions in tissue regeneration.
Collapse
Affiliation(s)
- Takehiro Yamazaki
- Faculty of Life and Medical Sciences, Doshisha University, Kyoto 610-0394, Japan.
| | - Toshifumi Kishimoto
- Faculty of Life and Medical Sciences, Doshisha University, Kyoto 610-0394, Japan.
| | - Paweł Leszczyński
- Institute of Genetics and Animal Breeding, Polish Academy of Sciences, Jastrzebiec, 05-552 Magdalenka, Poland.
| | - Koichiro Sadakane
- Faculty of Life and Medical Sciences, Doshisha University, Kyoto 610-0394, Japan.
| | - Takahiro Kenmotsu
- Faculty of Life and Medical Sciences, Doshisha University, Kyoto 610-0394, Japan.
| | - Hirofumi Watanabe
- Department of Functional Morphology, Division of Cell Regeneration and Transplantation, Nihon University School of Medicine, Tokyo 173-8610, Japan.
| | - Tomohiko Kazama
- Department of Functional Morphology, Division of Cell Regeneration and Transplantation, Nihon University School of Medicine, Tokyo 173-8610, Japan.
| | - Taro Matsumoto
- Department of Functional Morphology, Division of Cell Regeneration and Transplantation, Nihon University School of Medicine, Tokyo 173-8610, Japan.
| | - Kenichi Yoshikawa
- Faculty of Life and Medical Sciences, Doshisha University, Kyoto 610-0394, Japan.
| | - Hiroaki Taniguchi
- Institute of Genetics and Animal Breeding, Polish Academy of Sciences, Jastrzebiec, 05-552 Magdalenka, Poland.
| |
Collapse
|
32
|
Gullotta F, Izzo D, Scalera F, Palazzo B, Martin I, Sannino A, Gervaso F. Biomechanical evaluation of hMSCs-based engineered cartilage for chondral tissue regeneration. J Mech Behav Biomed Mater 2018; 86:294-304. [DOI: 10.1016/j.jmbbm.2018.06.040] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Revised: 06/18/2018] [Accepted: 06/25/2018] [Indexed: 01/22/2023]
|
33
|
Sun Y, Yan L, Chen S, Pei M. Functionality of decellularized matrix in cartilage regeneration: A comparison of tissue versus cell sources. Acta Biomater 2018; 74:56-73. [PMID: 29702288 PMCID: PMC7307012 DOI: 10.1016/j.actbio.2018.04.048] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Revised: 04/20/2018] [Accepted: 04/23/2018] [Indexed: 01/12/2023]
Abstract
Increasing evidence indicates that decellularized extracellular matrices (dECMs) derived from cartilage tissues (T-dECMs) or chondrocytes/stem cells (C-dECMs) can support proliferation and chondrogenic differentiation of cartilage-forming cells. However, few review papers compare the differences between these dECMs when they serve as substrates for cartilage regeneration. In this review, after an introduction of cartilage immunogenicity and decellularization methods to prepare T-dECMs and C-dECMs, a comprehensive comparison focuses on the effects of T-dECMs and C-dECMs on proliferation and chondrogenic differentiation of chondrocytes/stem cells in vitro and in vivo. Key factors within dECMs, consisting of microarchitecture characteristics and micromechanical properties as well as retained insoluble and soluble matrix components, are discussed in-depth for potential mechanisms underlying the functionality of these dECMs in regulating chondrogenesis. With this information, we hope to benefit dECM based cartilage engineering and tissue regeneration for future clinical application. STATEMENT OF SIGNIFICANCE The use of decellularized extracellular matrix (dECM) is becoming a promising approach for tissue engineering and regeneration. Compared to dECM derived from cartilage tissue, recently reported dECM from cell sources exhibits a distinct role in cell based cartilage regeneration. In this review paper, for the first time, tissue and cell based dECMs are comprehensively compared for their functionality in cartilage regeneration. This information is expected to provide an update for dECM based cartilage regeneration.
Collapse
Affiliation(s)
- Yu Sun
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV 26506, USA; Department of Orthopaedics, Orthopaedics Institute, Subei People's Hospital of Jiangsu Province, Yangzhou, Jiangsu 225001, China
| | - Lianqi Yan
- Department of Orthopaedics, Orthopaedics Institute, Subei People's Hospital of Jiangsu Province, Yangzhou, Jiangsu 225001, China
| | - Song Chen
- Department of Orthopaedics, Chengdu Military General Hospital, Chengdu, Sichuan 610083, China
| | - Ming Pei
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV 26506, USA; Exercise Physiology, West Virginia University, Morgantown, WV 26506, USA; WVU Cancer Institute, Robert C. Byrd Health Sciences Center, West Virginia University, Morgantown, WV 26506, USA.
| |
Collapse
|
34
|
Calabrese G, Gulino R, Giuffrida R, Forte S, Figallo E, Fabbi C, Salvatorelli L, Memeo L, Gulisano M, Parenti R. In Vivo Evaluation of Biocompatibility and Chondrogenic Potential of a Cell-Free Collagen-Based Scaffold. Front Physiol 2017; 8:984. [PMID: 29238307 PMCID: PMC5712864 DOI: 10.3389/fphys.2017.00984] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Accepted: 11/17/2017] [Indexed: 01/07/2023] Open
Abstract
Injured articular cartilage has a limited innate regenerative capacity, due to the avascular nature and low cellularity of the tissue itself. Although several approaches have been proposed to repair the joint cartilage, none of them has proven to be effective. The absence of suitable therapeutic options has encouraged tissue-engineering approaches combining specific cell types and biomaterials. In the present work, we have evaluated the potential of a cell-free Collagen I-based scaffold to promote the augmentation of cartilage-like phenotype after subcutaneous implantation in the mouse. Forty female mice were grafted subcutaneously with scaffolds, while four additional mice without scaffold were used as negative controls. The effects of scaffold were evaluated at 1, 2, 4, 8, or 16 weeks after implantation. Immunohistochemical analysis shows the expression of typical cartilage markers, including type-II Collagen, Aggrecan, Matrilin-1 and Sox 9. These data are also confirmed by qRT-PCR that further show that both COL2A1 and COL1A1 increase over time, but the first one increases more rapidly, thus suggesting a typical cartilage-like address. Histological analysis shows the presence of some pericellular lacunae, after 8 and 16 weeks. Results suggest that this scaffold (i) is biocompatible in vivo, (ii) is able to recruit host cells (iii) induce chondrogenic differentiation of host cells. Such evidences suggest that this cell-free scaffold is promising and represents a potential approach for cartilage regeneration.
Collapse
Affiliation(s)
- Giovanna Calabrese
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy.,Istituto Oncologico del Mediterraneo Ricerca, Catania, Italy
| | - Rosario Gulino
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy.,Istituto Oncologico del Mediterraneo Ricerca, Catania, Italy
| | | | - Stefano Forte
- Istituto Oncologico del Mediterraneo Ricerca, Catania, Italy
| | | | | | - Lucia Salvatorelli
- Department of Medical and Surgical Sciences and Advanced Technologies, G.F. Ingrassia, "Policlinico Vittorio Emanuele", Anatomic Pathology Section, University of Catania, Catania, Italy
| | - Lorenzo Memeo
- Department of Experimental Oncology, Mediterranean Institute of Oncology, Viagrande, Italy
| | - Massimo Gulisano
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Rosalba Parenti
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| |
Collapse
|
35
|
Biocompatibility of hydrogel-based scaffolds for tissue engineering applications. Biotechnol Adv 2017; 35:530-544. [DOI: 10.1016/j.biotechadv.2017.05.006] [Citation(s) in RCA: 407] [Impact Index Per Article: 50.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2016] [Revised: 05/08/2017] [Accepted: 05/22/2017] [Indexed: 12/15/2022]
|
36
|
Human adipose-derived mesenchymal stem cells seeded into a collagen-hydroxyapatite scaffold promote bone augmentation after implantation in the mouse. Sci Rep 2017; 7:7110. [PMID: 28769083 PMCID: PMC5541101 DOI: 10.1038/s41598-017-07672-0] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Accepted: 07/03/2017] [Indexed: 12/29/2022] Open
Abstract
Traumatic injury or surgical excision of diseased bone tissue usually require the reconstruction of large bone defects unable to heal spontaneously, especially in older individuals. This is a big challenge requiring the development of biomaterials mimicking the bone structure and capable of inducing the right commitment of cells seeded within the scaffold. In particular, given their properties and large availability, the human adipose-derived stem cells are considered as the better candidate for autologous cell transplantation. In order to evaluate the regenerative potential of these cells along with an osteoinductive biomaterial, we have used collagen/hydroxyapatite scaffolds to test ectopic bone formation after subcutaneous implantation in mice. The process was analysed both in vivo, by Fluorescent Molecular Tomography (FMT), and ex vivo, to evaluate the formation of bone and vascular structures. The results have shown that the biomaterial could itself be able of promoting differentiation of host cells and bone formation, probably by means of its intrinsic chemical and structural properties, namely the microenvironment. However, when charged with human mesenchymal stem cells, the ectopic bone formation within the scaffold was increased. We believe that these results represent an important advancement in the field of bone physiology, as well as in regenerative medicine.
Collapse
|
37
|
Yipeng J, Yongde X, Yuanyi W, Jilei S, Jiaxiang G, Jiangping G, Yong Y. Microtissues Enhance Smooth Muscle Differentiation and Cell Viability of hADSCs for Three Dimensional Bioprinting. Front Physiol 2017; 8:534. [PMID: 28790931 PMCID: PMC5524823 DOI: 10.3389/fphys.2017.00534] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Accepted: 07/10/2017] [Indexed: 12/22/2022] Open
Abstract
Smooth muscle differentiated human adipose derived stem cells (hADSCs) provide a crucial stem cell source for urinary tissue engineering, but the induction of hADSCs for smooth muscle differentiation still has several issues to overcome, including a relatively long induction time and equipment dependence, which limits access to abundant stem cells within a short period of time for further application. Three-dimensional (3D) bioprinting holds great promise in regenerative medicine due to its controllable construction of a designed 3D structure. When evenly mixed with bioink, stem cells can be spatially distributed within a bioprinted 3D structure, thus avoiding drawbacks such as, stem cell detachment in a conventional cell-scaffold strategy. Notwithstanding the advantages mentioned above, cell viability is often compromised during 3D bioprinting, which is often due to pressure during the bioprinting process. The objective of our study was to improve the efficiency of hADSC smooth muscle differentiation and cell viability of a 3D bioprinted structure. Here, we employed the hanging-drop method to generate hADSC microtissues in a smooth muscle inductive medium containing human transforming growth factor β1 and bioprinted the induced microtissues onto a 3D structure. After 3 days of smooth muscle induction, the expression of α-smooth muscle actin and smoothelin was higher in microtissues than in their counterpart monolayer cultured hADSCs, as confirmed by immunofluorescence and western blotting analysis. The semi-quantitative assay showed that the expression of α-smooth muscle actin (α-SMA) was 0.218 ± 0.077 in MTs and 0.082 ± 0.007 in Controls; smoothelin expression was 0.319 ± 0.02 in MTs and 0.178 ± 0.06 in Controls. Induced MTs maintained their phenotype after the bioprinting process. Live/dead and cell count kit 8 assays showed that cell viability and cell proliferation in the 3D structure printed with microtissues were higher at all time points compared to the conventional single-cell bioprinting strategy (mean cell viability was 88.16 ± 3.98 vs. 61.76 ± 15% for microtissues and single-cells, respectively). These results provide a novel way to enhance the smooth muscle differentiation of hADSCs and a simple method to maintain better cell viability in 3D bioprinting.
Collapse
Affiliation(s)
- Jin Yipeng
- Department of Urology, Chinese PLA General HospitalBeijing, China
| | - Xu Yongde
- Department of Urology, First Affiliated Hospital of Chinese PLA General HospitalBeijing, China
| | - Wu Yuanyi
- Department of Urology, First Affiliated Hospital of Chinese PLA General HospitalBeijing, China
| | - Sun Jilei
- Department of Urology, First Affiliated Hospital of Chinese PLA General HospitalBeijing, China
| | - Guo Jiaxiang
- Department of Urology, First Affiliated Hospital of Chinese PLA General HospitalBeijing, China
| | - Gao Jiangping
- Department of Urology, Chinese PLA General HospitalBeijing, China
| | - Yang Yong
- Department of Urology, First Affiliated Hospital of Chinese PLA General HospitalBeijing, China
| |
Collapse
|