scholarly journals In Vitro Screening of Molecularly Engineered Polyethylene Glycol Hydrogels for Cartilage Tissue Engineering using Periosteum-Derived and ATDC5 Cells

2018 ◽  
Vol 19 (11) ◽  
pp. 3341 ◽  
Author(s):  
Abhijith Kudva ◽  
Frank Luyten ◽  
Jennifer Patterson

The rapidly growing field of tissue engineering and regenerative medicine has brought about an increase in demand for biomaterials that mimic closely the form and function of biological tissues. Therefore, understanding the cellular response to the changes in material composition moves research one step closer to a successful tissue-engineered product. With this in mind, polyethylene glycol (PEG) hydrogels comprised of different concentrations of polymer (2.5%, 4%, 6.5%, or 8% (w/v)); different protease sensitive, peptide cross-linkers (VPMSMRGG or GPQGIWGQ); and the incorporation or lack of a peptide cell adhesion ligand (RGD) were screened for their ability to support in vitro chondrogenesis. Human periosteum-derived cells (hPDCs), a mesenchymal stem cell (MSC)-like primary cell source, and ATDC5 cells, a murine carcinoma-derived chondrogenic cell line, were encapsulated within the various hydrogels to assess the effects of the different formulations on cellular viability, proliferation, and chondrogenic differentiation while receiving exogenous growth factor stimulation via the medium. Through the results of this screening process, the 6.5% (w/v) PEG constructs, cross-linked with the GPQGIWGQ peptide and containing the RGD cell binding molecule, demonstrated an environment that consistently supported cellular viability and proliferation as well as chondrogenic differentiation.

2020 ◽  
Author(s):  
pengcheng xiao ◽  
Zhenglin Zhu ◽  
Chengcheng Du ◽  
Yongsheng Zeng ◽  
junyi Liao ◽  
...  

Abstract Background: Cartilage injuries pose formidable challenges for effective clinical management. Autologous stem cell-based therapies and transgene-enhanced cartilage tissue engineering may open new avenues for the treatment of cartilage injuries. Bone morphogenetic protein 2 (BMP2) is a promising chondrogenic growth factors for transgene-enhanced cartilage tissue engineering. However the BMP2 is failed to maintain a stable chondrogenic phenotype as it also induces robust endochondral ossification. Recently, human synovial derived mesenchymal stem cells (hSMSCs) arouse interested through the poor differentiation potential into osteogenic lineage. Smad7, a protein to antagonizes TGF-β/BMP signaling pathway has been discovered significant in the endochondral ossification. In the present study ,we further explore the effect of downregulate Smad7 in BMP2-induced chondrogenic differentiation of hSMSCs. Methods: hSMSCs were isolated from synovium of human knee joint through adhesion growth. In vitro and in vivo chondrogenic differentiation models of hSMSCs were constructed . Transgenes of BMP2, silencing Smad7 and Smad7 were expressed by adenoviral vectors. The osteogenic differentiation was detected by alkaline phosphatase staining, alizarin red staining. The chondrogenic differentiation was detected by alcian blue staining. Gene expression was determined by reverse transcription and quantitative real-time PCR (RT-qPCR), Immunofluorescence and immunohistochemistry. The subcutaneous stem cell implantation model was established and evaluated by micro-CT , h&e staining, alcian blue staining and immunohistochemistry assay.Results: Compared to other MSCs, hSMSCs performed less of capability to osteogenic differentiation. But the occurrence of endochondral ossification is still inevasible during BMP2 induced cartilage formation. We found that silencing Smad7 enhanced the BMP2-induced chondrogenic differentiation of hSMSCs in vitro. Also, it leading to much less of hypertrophic differentiation. The subcutaneous stem cells implantation assays demonstrated silencing Smad7 potentiates BMP2-induced cartilage formation and inhibits endochondral ossification. Conclusion: This study strongly suggests that application of hSMSCs , cell scaffolds and silencing Smad7 can potentiate BMP2-induced chondrogenic differentiation and inhibit endochondral ossification. Thus, inhibit the expression of Smad7 in BMP2-induced hSMSCs differentiation may be a new strategy for cartilage tissue engineering.


2021 ◽  
pp. 088532822110448
Author(s):  
Xiang Zhang ◽  
Zhenhao Yan ◽  
Guotao Guan ◽  
Zijing Lu ◽  
Shujie Yan ◽  
...  

Natural cartilage tissue has excellent mechanical properties and has certain cellular components. At this stage, it is a great challenge to produce cartilage scaffolds with excellent mechanical properties, biocompatibility, and biodegradability. Hydrogels are commonly used in tissue engineering because of their excellent biocompatibility; however, the mechanical properties of commonly used hydrogels are difficult to meet the requirements of making cartilage scaffolds. The mechanical properties of high concentration polyethylene glycol diacrylate (PEGDA) hydrogel are similar to those of natural cartilage, but its biocompatibility is poor. Low concentration hydrogel has better biocompatibility, but its mechanical properties are poor. In this study, two different hydrogels were combined to produce cartilage scaffolds with good mechanical properties and strong biocompatibility. First, the PEGDA grid scaffold was printed with light curing 3D printing technology, and then the low concentration GelMA/Alginate hydrogel with chondral cells was filled into the PEGDA grid scaffold. After a series of cell experiments, the filling hydrogel with the best biocompatibility was screened out, and finally the filled hydrogel with cells and excellent biocompatibility was obtained. Cartilage tissue engineering scaffolds with certain mechanical properties were found to have a tendency of cartilage formation in in vitro culture. Compared with the scaffold obtained by using a single hydrogel, this molding method can produce a tissue engineering scaffold with excellent mechanical properties on the premise of ensuring biocompatibility, which has a certain potential application value in the field of cartilage tissue engineering.


Biomolecules ◽  
2021 ◽  
Vol 11 (1) ◽  
pp. 115
Author(s):  
Ching-Yun Chen ◽  
Chunching Li ◽  
Cherng-Jyh Ke ◽  
Jui-Sheng Sun ◽  
Feng-Huei Lin

Human cartilage has relatively slow metabolism compared to other normal tissues. Cartilage damage is of great clinical consequence since cartilage has limited intrinsic healing potential. Cartilage tissue engineering is a rapidly emerging field that holds great promise for tissue function repair and artificial/engineered tissue substitutes. However, current clinical therapies for cartilage repair are less than satisfactory and rarely recover full function or return the diseased tissue to its native healthy state. Kartogenin (KGN), a small molecule, can promote chondrocyte differentiation both in vitro and in vivo. The purpose of this research is to optimize the chondrogenic process in mesenchymal stem cell (MSC)-based chondrogenic constructs with KGN for potential use in cartilage tissue engineering. In this study, we demonstrate that KGN treatment can promote MSC condensation and cell cluster formation within a tri-copolymer scaffold. Expression of Acan, Sox9, and Col2a1 was significantly up-regulated in three-dimensional (3D) culture conditions. The lacuna-like structure showed active deposition of type II collagen and aggrecan deposition. We expect these results will open new avenues for the use of small molecules in chondrogenic differentiation protocols in combination with scaffolds, which may yield better strategies for cartilage tissue engineering.


2018 ◽  
Vol 8 (8) ◽  
pp. 1398 ◽  
Author(s):  
Shohei Ishikawa ◽  
Kazutoshi Iijima ◽  
Kohei Sasaki ◽  
Mineo Hashizume ◽  
Masaaki Kawabe ◽  
...  

In cartilage tissue engineering, three-dimensional (3D) scaffolds provide native extracellular matrix (ECM) environments that induce tissue ingrowth and ECM deposition for in vitro and in vivo tissue regeneration. In this report, we investigated 3D silica nonwoven fabrics (Cellbed®) as a scaffold for mesenchymal stem cells (MSCs) in cartilage tissue engineering applications. The unique, highly porous microstructure of 3D silica fabrics allows for immediate cell infiltration for tissue repair and orientation of cell–cell interaction. It is expected that the morphological similarity of silica fibers to that of fibrillar ECM contributes to the functionalization of cells. Human bone marrow-derived MSCs were cultured in 3D silica fabrics, and chondrogenic differentiation was induced by culture in chondrogenic differentiation medium. The characteristics of chondrogenic differentiation including cellular growth, ECM deposition of glycosaminoglycan and collagen, and gene expression were evaluated. Because of the highly interconnected network structure, stiffness, and permeability of the 3D silica fabrics, the level of chondrogenesis observed in MSCs seeded within was comparable to that observed in MSCs maintained on atelocollagen gels, which are widely used to study the chondrogenesis of MSCs in vitro and in vivo. These results indicated that 3D silica nonwoven fabrics are a promising scaffold for the regeneration of articular cartilage defects using MSCs, showing the particular importance of high elasticity.


Biomaterials ◽  
2011 ◽  
Vol 32 (25) ◽  
pp. 5773-5781 ◽  
Author(s):  
Nandana Bhardwaj ◽  
Quynhhoa T. Nguyen ◽  
Albert C. Chen ◽  
David L. Kaplan ◽  
Robert L. Sah ◽  
...  

2016 ◽  
Vol 2016 ◽  
pp. 1-12 ◽  
Author(s):  
Stefano Focaroli ◽  
Gabriella Teti ◽  
Viviana Salvatore ◽  
Isabella Orienti ◽  
Mirella Falconi

Articular cartilage is a highly organized tissue with complex biomechanical properties. However, injuries to the cartilage usually lead to numerous health concerns and often culminate in disabling symptoms, due to the poor intrinsic capacity of this tissue for self-healing. Although various approaches are proposed for the regeneration of cartilage, its repair still represents an enormous challenge for orthopedic surgeons. The field of tissue engineering currently offers some of the most promising strategies for cartilage restoration, in which assorted biomaterials and cell-based therapies are combined to develop new therapeutic regimens for tissue replacement. The current study describes thein vitrobehavior of human adipose-derived mesenchymal stem cells (hADSCs) encapsulated within calcium/cobalt (Ca/Co) alginate beads. These novel chondrogenesis-promoting scaffolds take advantage of the synergy between the alginate matrix and Co+2ions, without employing costly growth factors (e.g., transforming growth factor betas (TGF-βs) or bone morphogenetic proteins (BMPs)) to direct hADSC differentiation into cartilage-producing chondrocytes.


2021 ◽  
Author(s):  
Anggraini Barlian ◽  
Dinda Hani’ah Arum Saputri ◽  
Adriel Hernando ◽  
Ekavianty Prajatelistia ◽  
Hutomo Tanoto

Abstract Cartilage tissue engineering, particularly micropattern, can influence the biophysical properties of mesenchymal stem cells (MSCs) leading to chondrogenesis. In this research, human Wharton’s jelly MSCs (hWJ-MSCs) were grown on a striped micropattern containing spider silk protein (spidroin) from Argiope appensa. This research aims to direct hWJ-MSCs chondrogenesis using micropattern made of spidroin bioink as opposed to fibronectin that often used as the gold standard. Cells were cultured on striped micropattern of 500 µm and 1000 µm width sizes without chondrogenic differentiation medium for 21 days. The immunocytochemistry result showed that spidroin contains RGD sequences and facilitates cell adhesion via integrin β1. Chondrogenesis was observed through the expression of glycosaminoglycan, type II collagen, and SOX9. The result on glycosaminoglycan content proved that 1000 µm was the optimal width to support chondrogenesis. Spidroin micropattern induced significantly higher expression of SOX9 mRNA on day-21 and SOX9 protein was located inside the nucleus starting from day-7. COL2A1 mRNA of spidroin micropattern groups was downregulated on day-21 and collagen type II protein was detected starting from day-14. These results showed that spidroin micropattern enhances chondrogenic markers while maintains long-term upregulation of SOX9, and therefore has the potential as a new method for cartilage tissue engineering.


2009 ◽  
Vol 21 (03) ◽  
pp. 149-155 ◽  
Author(s):  
Hsu-Wei Fang

Cartilage injuries may be caused by trauma, biomechanical imbalance, or degenerative changes of joint. Unfortunately, cartilage has limited capability to spontaneous repair once damaged and may lead to progressive damage and degeneration. Cartilage tissue-engineering techniques have emerged as the potential clinical strategies. An ideal tissue-engineering approach to cartilage repair should offer good integration into both the host cartilage and the subchondral bone. Cells, scaffolds, and growth factors make up the tissue engineering triad. One of the major challenges for cartilage tissue engineering is cell source and cell numbers. Due to the limitations of proliferation for mature chondrocytes, current studies have alternated to use stem cells as a potential source. In the recent years, a lot of novel biomaterials has been continuously developed and investigated in various in vitro and in vivo studies for cartilage tissue engineering. Moreover, stimulatory factors such as bioactive molecules have been explored to induce or enhance cartilage formation. Growth factors and other additives could be added into culture media in vitro, transferred into cells, or incorporated into scaffolds for in vivo delivery to promote cellular differentiation and tissue regeneration.Based on the current development of cartilage tissue engineering, there exist challenges to overcome. How to manipulate the interactions between cells, scaffold, and signals to achieve the moderation of implanted composite differentiate into moderate stem cells to differentiate into hyaline cartilage to perform the optimum physiological and biomechanical functions without negative side effects remains the target to pursue.


Sign in / Sign up

Export Citation Format

Share Document