Displaying publications 81 - 100 of 165 in total

Abstract:
Sort:
  1. Halib N, Ahmad I, Grassi M, Grassi G
    Int J Pharm, 2019 Jul 20;566:631-640.
    PMID: 31195074 DOI: 10.1016/j.ijpharm.2019.06.017
    Cellulose is a natural homopolymer, composed of β-1,4- anhydro-d-glucopyranose units. Unlike plant cellulose, bacterial cellulose (BC), obtained from species belonging to the genera of Acetobacter, Rhizobium, Agrobacterium, and Sarcina through various cultivation methods and techniques, is produced in its pure form. BC is produced in the form of gel-like, never dry sheet with tremendous mechanical properties. Containing up to 99% of water, BC hydrogel is considered biocompatible thus finding robust applications in the health industry. Moreover, BC three-dimensional structure closely resembles the extracellular matrix (ECM) of living tissue. In this review, we focus on the porous BC morphology particularly suited to host oxygen and nutrients thus providing conducive environment for cell growth and proliferation. The remarkable BC porous morphology makes this biological material a promising templet for the generation of 3D tissue culture and possibly for tissue-engineered scaffolds.
    Matched MeSH terms: Tissue Scaffolds
  2. Sukmana I
    J Artif Organs, 2012 Sep;15(3):215-24.
    PMID: 22527978 DOI: 10.1007/s10047-012-0644-6
    Tissue engineering seeks strategies to design polymeric scaffolds that allow high-cell-density cultures with signaling molecules and suitable vascular supply. One major obstacle in tissue engineering is the inability to create thick engineered-tissue constructs. A pre-vascularized tissue scaffold appears to be the most favorable approach to avoid nutrient and oxygen supply limitations as well as to allow waste removal, factors that are often hurdles in developing thick engineered tissues. Vascularization can be achieved using strategies in which cells are cultured in bioactive polymer scaffolds that can mimic extracellular matrix environments. This review addresses recent advances and future challenges in developing and using bioactive polymer scaffolds to promote tissue construct vascularization.
    Matched MeSH terms: Tissue Scaffolds*
  3. Zulkifli FH, Jahir Hussain FS, Abdull Rasad MS, Mohd Yusoff M
    J Biomater Appl, 2015 Feb;29(7):1014-27.
    PMID: 25186524 DOI: 10.1177/0885328214549818
    The aim of this research is to develop biocompatible nanofibrous mats using hydroxyethyl cellulose with improved cellular adhesion profiles and stability and use these fibrous mats as potential scaffold for skin tissue engineering. Glutaraldehyde was used to treat the scaffolds water insoluble as well as improve their biostability for possible use in biomedical applications. Electrospinning of hydroxyethyl cellulose (5 wt%) with poly(vinyl alcohol) (15 wt%) incorporated with and without collagen was blended at (1:1:1) and (1:1) ratios, respectively, and was evaluated for optimal criteria as tissue engineering scaffolds. The nanofibrous mats were crosslinked and characterized by scanning electron microscope, Fourier transform infrared spectroscopy, differential scanning calorimetry, and thermogravimetric analysis. Scanning electron microscope images showed that the mean diameters of blend nanofibers were gradually increased after chemically crosslinking with glutaraldehyde. Fourier transform infrared spectroscopy was carried out to understand chemical interactions in the presence of aldehyde groups. Thermal characterization results showed that the stability of hydroxyethyl cellulose/poly(vinyl alcohol) and hydroxyethyl cellulose/poly(vinyl alcohol)/collagen nanofibers was increased with glutaraldehyde treatment. Studies on cell-scaffolds interaction were carried out by culturing human fibroblast (hFOB) cells on the nanofibers by assessing the growth, proliferation, and morphologies of cells. The scanning electron microscope results show that better cell proliferation and attachment appeared on hydroxyethyl cellulose/poly(vinyl alcohol)/collagen substrates after 7 days of culturing, thus, promoting the potential of electrospun scaffolds as a promising candidate for tissue engineering applications.
    Matched MeSH terms: Tissue Scaffolds/chemistry*
  4. Azhim A, Syazwani N, Morimoto Y, Furukawa KS, Ushida T
    J Biomater Appl, 2014 Jul;29(1):130-41.
    PMID: 24384523 DOI: 10.1177/0885328213517579
    A novel decellularization method using sonication treatment is described. Sonication treatment is the combination of physical and chemical agents. These methods will disrupt cell membrane and release cell contents to external environments. The cell removal was facilitated by subsequent rinsing of sodium dodecyl sulfate detergents. Sonication treatment is used in the preparation of complete decellularized bioscaffolds. The aim of this study is to confirm the usefulness of sonication treatment for preparation of biological scaffolds. In this study, samples of aortic tissues are decellularized by sonication treatment at frequency of 170 kHz in 0.1% and 2% sodium dodecyl sulfate detergents for 10-h treatment time. The relation between decellularization and sonication parameters such as dissolved oxygen concentration, conductivity, and pH is investigated. Histological analysis and biomechanical testing is performed to evaluate cell removal efficiency as well as changes in biomechanical properties. Minimal inflammation response elicit by bioscaffolds is confirmed by xenogeneic implantation and immunohistochemistry. Sonication treatment is able to produce complete decellularized tissue suggesting that these treatments could be applied widely as one of the decellularization method.
    Matched MeSH terms: Tissue Scaffolds*
  5. Lew KS, Othman R, Ishikawa K, Yeoh FY
    J Biomater Appl, 2012 Sep;27(3):345-58.
    PMID: 21862511 DOI: 10.1177/0885328211406459
    This review summarises the major developments of macroporous bioceramics used mainly for repairing bone defects. Porous bioceramics have been receiving attention ever since their larger surface area was reported to be beneficial for the formation of more rigid bonds with host tissues. The study of porous bioceramics is important to overcome the less favourable bonds formed between dense bioceramics and host tissues, especially in healing bone defects. Macroporous bioceramics, which have been studied extensively, include hydroxyapatite, tricalcium phosphate, alumina, and zirconia. The pore size and interconnections both have significant effects on the growth rate of bone tissues. The optimum pore size of hydroxyapatite scaffolds for bone growth was found to be 300 µm. The existence of interconnections between pores is critical during the initial stage of tissue ingrowth on porous hydroxyapatite scaffolds. Furthermore, pore formation on β-tricalcium phosphate scaffolds also allowed the impregnation of growth factors and cells to improve bone tissues growth significantly. The formation of vascularised tissues was observed on macroporous alumina but did not take place in the case of dense alumina due to its bioinert nature. A macroporous alumina coating on scaffolds was able to improve the overall mechanical properties, and it enabled the impregnation of bioactive materials that could increase the bone growth rate. Despite the bioinertness of zirconia, porous zirconia was useful in designing scaffolds with superior mechanical properties after being coated with bioactive materials. The pores in zirconia were believed to improve the bone growth on the coated system. In summary, although the formation of pores in bioceramics may adversely affect mechanical properties, the advantages provided by the pores are crucial in repairing bone defects.
    Matched MeSH terms: Tissue Scaffolds
  6. Latfi ASA, Pramanik S, Poon CT, Gumel AM, Lai KW, Annuar MSM, et al.
    J Biomater Appl, 2019 01;33(6):854-865.
    PMID: 30458659 DOI: 10.1177/0885328218812490
    Natural biopolymers have many attractive medical applications; however, complications due to fibrosis caused a reduction in diffusion and dispersal of nutrients and waste products. Consequently, severe immunocompatibility problems and poor mechanical and degradation properties in synthetic polymers ensue. Hence, the present study investigates a novel hydrogel material synthesized from caprolactone, ethylene glycol, ethylenediamine, polyethylene glycol, ammonium persulfate, and tetramethylethylenediamine via chemo-enzymatic route. Spectroscopic analyses indicated the formation of polyurea and polyhydroxyurethane as the primary building block of the hydrogel starting material. Biocompatibility studies showed positive observation in biosafety test using direct contact cytotoxicity assay in addition to active cellular growth on the hydrogel scaffold based on fluorescence observation. The synthesized hydrogel also exhibited (self)fluorescence properties under specific wavelength excitation. Hence, synthesized hydrogel could be a potential candidate for medical imaging as well as tissue engineering applications as a tissue expander, coating material, biosensor, and drug delivery system.
    Matched MeSH terms: Tissue Scaffolds/chemistry
  7. Ching KY, Andriotis O, Sengers B, Stolz M
    J Biomater Appl, 2021 09;36(3):503-516.
    PMID: 33730922 DOI: 10.1177/08853282211002015
    Towards optimizing the growth of extracellular matrix to produce repair cartilage for healing articular cartilage (AC) defects in joints, scaffold-based tissue engineering approaches have recently become a focus of clinical research. Scaffold-based approaches by electrospinning aim to support the differentiation of chondrocytes by providing an ultrastructure similar to the fibrillar meshwork in native cartilage. In a first step, we demonstrate how the blending of chitosan with poly(ethylene oxide) (PEO) allows concentrated chitosan solution to become electrospinnable. The chitosan-based scaffolds share the chemical structure and characteristics of glycosaminoglycans, which are important structural components of the cartilage extracellular matrix. Electrospinning produced nanofibrils of ∼100 nm thickness that are closely mimicking the size of collagen fibrils in human AC. The polymer scaffolds were stabilized in physiological conditions and their stiffness was tuned by introducing the biocompatible natural crosslinker genipin. We produced scaffolds that were crosslinked with 1.0% genipin to obtain values of stiffness that were in between the stiffness of the superficial zone human AC of 600 ± 150 kPa and deep zone AC of 1854 ± 483 kPa, whereas the stiffness of 1.5% genipin crosslinked scaffold was similar to the stiffness of deep zone AC. The scaffolds were degradable, which was indicated by changes in the fibril structure and a decrease in the scaffold stiffness after seven months. Histological and immunohistochemical analysis after three weeks of culture with human articular chondrocytes (HACs) showed a cell viability of over 90% on the scaffolds and new extracellular matrix deposited on the scaffolds.
    Matched MeSH terms: Tissue Scaffolds/chemistry
  8. Nazemi N, Rajabi N, Aslani Z, Kharaziha M, Kasiri-Asgarani M, Bakhsheshi-Rad HR, et al.
    J Biomater Appl, 2023 Jan;37(6):979-991.
    PMID: 36454961 DOI: 10.1177/08853282221140672
    Porous structure, biocompatibility and biodegradability, large surface area, and drug-loading ability are some remarkable properties of zeolite structure, making it a great possible option for bone tissue engineering. Herein, we evaluated the potential application of the ZSM-5 scaffold encapsulated GEN with high porosity structure and significant antibacterial properties. The space holder process has been employed as a new fabrication method with interconnected pores and suitable mechanical properties. In this study, for the first time, ZSM-5 scaffolds with GEN drug-loading were fabricated with the space holder method. The results showed excellent open porosity in the range of 70-78% for different GEN concentrations and appropriate mechanical properties. Apatite formation on the scaffold surface was determined with Simulation body fluid (SBF), and a new bone-like apatite layer shaping on all samples confirmed the in vitro bioactivity of ZSM-5-GEN scaffolds. Also, antibacterial properties were investigated against both gram-positive and gram-negative bacteria. The incorporation of various amounts of GEN increased the inhibition zone from 24 to 28 (for E. coli) and 26 to 37 (for S. aureus). In the culture with MG63 cells, great cell viability and high cell proliferation after 7 days of culture were determined.
    Matched MeSH terms: Tissue Scaffolds/chemistry
  9. Sha'ban M, Yoon SJ, Ko YK, Ha HJ, Kim SH, So JW, et al.
    J Biomater Sci Polym Ed, 2008;19(9):1219-37.
    PMID: 18727862 DOI: 10.1163/156856208785540163
    Previously, we have proven that fibrin and poly(lactic-co-glycolic acid) (PLGA) scaffolds facilitate cell proliferation, matrix production and early chondrogenesis of rabbit articular chondrocytes in in vitro and in vivo experiments. In this study, we evaluated the potential of fibrin/PLGA scaffold for intervertebral disc (IVD) tissue engineering using annulus fibrosus (AF) and nucleus pulposus (NP) cells in relation to potential clinical application. PLGA scaffolds were soaked in cells-fibrin suspension and polymerized by dropping thrombin-sodium chloride (CaCl(2)) solution. A PLGA-cell complex without fibrin was used as control. Higher cellular proliferation activity was observed in fibrin/PLGA-seeded AF and NP cells at each time point of 3, 7, 14 and 7 days using the MTT assay. After 3 weeks in vitro incubation, fibrin/PLGA exhibited a firmer gross morphology than PLGA groups. A significant cartilaginous tissue formation was observed in fibrin/PLGA, as proven by the development of cells cluster of various sizes and three-dimensional (3D) cartilaginous histoarchitecture and the presence of proteoglycan-rich matrix and glycosaminoglycan (GAG). The sGAG production measured by 1,9-dimethylmethylene blue (DMMB) assay revealed greater sGAG production in fibrin/PLGA than PLGA group. Immunohistochemical analyses showed expressions of collagen type II, aggrecan core protein and collagen type I genes throughout in vitro culture in both fibrin/PLGA and PLGA. In conclusion, fibrin promotes cell proliferation, stable in vitro tissue morphology, superior cartilaginous tissue formation and sGAG production of AF and NP cells cultured in PLGA scaffold. The 3D porous PLGA scaffold-cell complexes using fibrin can provide a vehicle for delivery of cells to regenerate tissue-engineered IVD tissue.
    Matched MeSH terms: Tissue Scaffolds/chemistry*
  10. Boukari Y, Scurr DJ, Qutachi O, Morris AP, Doughty SW, Rahman CV, et al.
    J Biomater Sci Polym Ed, 2015;26(12):796-811.
    PMID: 26065672 DOI: 10.1080/09205063.2015.1058696
    An injectable poly(DL-lactic-co-glycolic acid) (PLGA) system comprising both porous and protein-loaded microspheres capable of forming porous scaffolds at body temperature was developed for tissue regeneration purposes. Porous and non-porous (lysozyme loaded) PLGA microspheres were formulated to represent 'low molecular weight' 22-34 kDa, 'intermediate molecular weight' (IMW) 53 kDa and 'high molecular weight' 84-109 kDa PLGA microspheres. The respective average size of the microspheres was directly related to the polymer molecular weight. An initial burst release of lysozyme was observed from both microspheres and scaffolds on day 1. In the case of the lysozyme-loaded microspheres, this burst release was inversely related to the polymer molecular weight. Similarly, scaffolds loaded with 1 mg lysozyme/g of scaffold exhibited an inverse release relationship with polymer molecular weight. The burst release was highest amongst IMW scaffolds loaded with 2 and 3 mg/g. Sustained lysozyme release was observed after day 1 over 50 days (microspheres) and 30 days (scaffolds). The compressive strengths of the scaffolds were found to be inversely proportional to PLGA molecular weight at each lysozyme loading. Surface analysis indicated that some of the loaded lysozyme was distributed on the surfaces of the microspheres and thus responsible for the burst release observed. Overall the data demonstrates the potential of the scaffolds for use in tissue regeneration.
    Matched MeSH terms: Tissue Scaffolds/chemistry*
  11. Gobinathan S, Zainol SS, Azizi SF, Iman NM, Muniandy R, Hasmad HN, et al.
    J Biomater Sci Polym Ed, 2018 12;29(17):2051-2067.
    PMID: 29983100 DOI: 10.1080/09205063.2018.1485814
    Amniotic membrane has the potential to be used as scaffold in various tissue engineering applications. However, increasing its biostability at the same time maintaining its biocompatibility is important to enhance its usage as a scaffold. This studied characteristics genipin-crosslinked amniotic membrane as a bioscaffold. Redundant human amniotic membranes (HAM) divided into native (nAM), decellularized (dAM) and genipin-crosslinked (clAM) groups. The dAM and clAM group were decellularized using thermolysin (TL) and sodium hydroxide (NaOH) solution. Next, clAM group was crosslinked with 0.5% and 1.0% (w/v) genipin. The HAM was then studied for in vitro degradation, percentage of swelling, optical clarity, ultrastructure and mechanical strength. Meanwhile, fibroblasts isolated from nasal turbinates were then seeded onto nAM, dAM and clAM for biocompatibility studies. clAM had the slowest degradation rate and were still morphologically intact after 30 days of incubation in 0.01% collagenase type 1 solution. The dAM had a significantly highest percentage of swelling than other groups (p 
    Matched MeSH terms: Tissue Scaffolds/chemistry*
  12. Boukari Y, Qutachi O, Scurr DJ, Morris AP, Doughty SW, Billa N
    J Biomater Sci Polym Ed, 2017 Nov;28(16):1966-1983.
    PMID: 28777694 DOI: 10.1080/09205063.2017.1364100
    The development of patient-friendly alternatives to bone-graft procedures is the driving force for new frontiers in bone tissue engineering. Poly (dl-lactic-co-glycolic acid) (PLGA) and chitosan are well-studied and easy-to-process polymers from which scaffolds can be fabricated. In this study, a novel dual-application scaffold system was formulated from porous PLGA and protein-loaded PLGA/chitosan microspheres. Physicochemical and in vitro protein release attributes were established. The therapeutic relevance, cytocompatibility with primary human mesenchymal stem cells (hMSCs) and osteogenic properties were tested. There was a significant reduction in burst release from the composite PLGA/chitosan microspheres compared with PLGA alone. Scaffolds sintered from porous microspheres at 37 °C were significantly stronger than the PLGA control, with compressive strengths of 0.846 ± 0.272 MPa and 0.406 ± 0.265 MPa, respectively (p 
    Matched MeSH terms: Tissue Scaffolds/chemistry*
  13. Yousefi AM, Hoque ME, Prasad RG, Uth N
    J Biomed Mater Res A, 2015 Jul;103(7):2460-81.
    PMID: 25345589 DOI: 10.1002/jbm.a.35356
    The repair of osteochondral defects requires a tissue engineering approach that aims at mimicking the physiological properties and structure of two different tissues (cartilage and bone) using specifically designed scaffold-cell constructs. Biphasic and triphasic approaches utilize two or three different architectures, materials, or composites to produce a multilayered construct. This article gives an overview of some of the current strategies in multiphasic/gradient-based scaffold architectures and compositions for tissue engineering of osteochondral defects. In addition, the application of finite element analysis (FEA) in scaffold design and simulation of in vitro and in vivo cell growth outcomes has been briefly covered. FEA-based approaches can potentially be coupled with computer-assisted fabrication systems for controlled deposition and additive manufacturing of the simulated patterns. Finally, a summary of the existing challenges associated with the repair of osteochondral defects as well as some recommendations for future directions have been brought up in the concluding section of this article.
    Matched MeSH terms: Tissue Scaffolds*
  14. Krishnamurithy G, Shilpa PN, Ahmad RE, Sulaiman S, Ng CL, Kamarul T
    J Biomed Mater Res A, 2011 Dec 01;99(3):500-6.
    PMID: 21913317 DOI: 10.1002/jbm.a.33184
    Human amniotic membrane (HAM) is an established biomaterial used in many clinical applications. However, its use for tissue engineering purposes has not been fully realized. A study was therefore conducted to evaluate the feasibility of using HAM as a chondrocyte substrate/carrier. HAMs were obtained from fresh human placenta and were process to produced air dried HAM (AdHAM) and freeze dried HAM (FdHAM). Rabbit chondrocytes were isolated and expanded in vitro and seeded onto these preparations. Cell proliferation, GAG expression and GAG/cell expression were measured at days 3, 6, 9, 12, 15, 21, and 28. These were compared to chondrocytes seeded onto plastic surfaces. Histological analysis and scanning electron microscopy was performed to observe cell attachment. There was significantly higher cell proliferation rates observed between AdHAM (13-51%, P=0.001) or FdHAM (18-48%, p = 0.001) to chondrocytes in monolayer. Similarly, GAG and GAG/cell expressed in AdHAM (33-82%, p = 0.001; 22-60%, p = 0.001) or FdHAM (41-81%, p = 0.001: 28-60%, p = 0.001) were significantly higher than monolayer cultures. However, no significant differences were observed in the proliferation rates (p = 0.576), GAG expression (p = 0.476) and GAG/cell expression (p = 0.135) between AdHAM and FdHAM. The histology and scanning electron microscopy assessments demonstrates good chondrocyte attachments on both HAMs. In conclusion, both AdHAM and FdHAM provide superior chondrocyte proliferation, GAG expression, and attachment than monolayer cultures making it a potential substrate/carrier for cell based cartilage therapy and transplantation.
    Matched MeSH terms: Tissue Scaffolds/chemistry*
  15. Foo LH, Suzina AH, Azlina A, Kannan TP
    J Biomed Mater Res A, 2008 Oct;87(1):215-21.
    PMID: 18085658
    Coral matrix of Porites sp. has the suitable properties for bone cell growth. This study was aimed to study the gene expression levels of osteoblast specific genetic markers; RUNX2, osteopontin, alkaline phosphatase and osteocalcin from osteoblasts seeded in coral scaffold, which are important in determining the feasibility of osteoblasts. Human osteoblasts were inoculated onto the processed coral in Dulbecco's Minimum Essential Medium. The cells were trypsinized on day 1, 7, 14, 18, and 21 and added with RNALater for preservation of RNA in cells. The RNA was extracted using commercial RNA extraction kit and the respective genes were amplified using RT-PCR kit and analyzed qualitatively on 1.5% agarose gel. The expressions were evaluated with the Integrated Density Value based on the intensity of band for different periods of cell harvest. Increased expressions of the RUNX2, osteopontin, alkaline phosphatase and osteocalcin genes in the present study proved that coral is a favorable carrier for osteogenetically competent cells to attach and remain viable.
    Matched MeSH terms: Tissue Scaffolds*
  16. Salem SA, Hwei NM, Bin Saim A, Ho CC, Sagap I, Singh R, et al.
    J Biomed Mater Res A, 2013 Aug;101(8):2237-47.
    PMID: 23349110 DOI: 10.1002/jbm.a.34518
    The chief obstacle for reconstructing the bladder is the absence of a biomaterial, either permanent or biodegradable, that will function as a suitable scaffold for the natural process of regeneration. In this study, polylactic-co-glycolic acid (PLGA) plus collagen or fibrin was evaluated for its suitability as a scaffold for urinary bladder construct. Human adipose-derived stem cells (HADSCs) were cultured, followed by incubation in smooth muscle cells differentiation media. Differentiated HADSCs were then seeded onto PLGA mesh supported with collagen or fibrin. Evaluation of cell-seeded PLGA composite immersed in culture medium was performed under a light and scanning microscope. To determine if the composite is compatible with the urodynamic properties of urinary bladder, porosity and leaking test was performed. The PLGA samples were subjected to tensile testing was pulled until PLGA fibers break. The results showed that the PLGA composite is biocompatible to differentiated HADSCs. PLGA-collagen mesh appeared to be optimal as a cell carrier while the three-layered PLGA-fibrin composite is better in relation to its leaking/ porosity property. A biomechanical test was also performed for three-layered PLGA with biological adhesive and three-layered PLGA alone. The tensile stress at failure was 30.82 ± 3.80 (MPa) and 34.36 ± 2.57 (MPa), respectively. Maximum tensile strain at failure was 19.42 ± 2.24 (mm) and 23.06 ± 2.47 (mm), respectively. Young's modulus was 0.035 ± 0.0083 and 0.043 ± 0.012, respectively. The maximum load at break was 58.55 ± 7.90 (N) and 65.29 ± 4.89 (N), respectively. In conclusion, PLGA-Fibrin fulfils the criteria as a scaffold for urinary bladder reconstruction.
    Matched MeSH terms: Tissue Scaffolds/chemistry*
  17. Rozila I, Azari P, Munirah S, Wan Safwani WK, Gan SN, Nur Azurah AG, et al.
    J Biomed Mater Res A, 2016 Feb;104(2):377-87.
    PMID: 26414782 DOI: 10.1002/jbm.a.35573
    The osteogenic potential of human adipose-derived stem cells (HADSCs) co-cultured with human osteoblasts (HOBs) using selected HADSCs/HOBs ratios of 1:1, 2:1, and 1:2, respectively, is evaluated. The HADSCs/HOBs were seeded on electrospun three-dimensional poly[(R)-3-hydroxybutyric acid] (PHB) blended with bovine-derived hydroxyapatite (BHA). Monocultures of HADSCs and HOBs were used as control groups. The effects of PHB-BHA scaffold on cell proliferation and cell morphology were assessed by AlamarBlue assay and field emission scanning electron microscopy. Cell differentiation, cell mineralization, and osteogenic-related gene expression of co-culture HADSCs/HOBs were examined by alkaline phosphatase (ALP) assay, alizarin Red S assay, and quantitative real time PCR, respectively. The results showed that co-culture of HADSCs/HOBs, 1:1 grown into PHB-BHA promoted better cell adhesion, displayed a significant higher cell proliferation, higher production of ALP, extracellular mineralization and osteogenic-related gene expression of run-related transcription factor, bone sialoprotein, osteopontin, and osteocalcin compared to other co-culture groups. This result also suggests that the use of electrospun PHB-BHA in a co-culture HADSCs/HOBs system may serve as promising approach to facilitate osteogenic differentiation activity of HADSCs through direct cell-to-cell contact with HOBs.
    Matched MeSH terms: Tissue Scaffolds/chemistry*
  18. Rizwan M, Hamdi M, Basirun WJ
    J Biomed Mater Res A, 2017 Nov;105(11):3197-3223.
    PMID: 28686004 DOI: 10.1002/jbm.a.36156
    Bioglass® 45S5 (BG) has an outstanding ability to bond with bones and soft tissues, but its application as a load-bearing scaffold material is restricted due to its inherent brittleness. BG-based composites combine the amazing biological and bioactive characteristics of BG with structural and functional features of other materials. This article reviews the composites of Bioglass® in combination with metals, ceramics and polymers for a wide range of potential applications from bone scaffolds to nerve regeneration. Bioglass® also possesses angiogenic and antibacterial properties in addition to its very high bioactivity; hence, composite materials developed for these applications are also discussed. BG-based composites with polymer matrices have been developed for a wide variety of soft tissue engineering. This review focuses on the research that suggests the suitability of BG-based composites as a scaffold material for hard and soft tissues engineering. Composite production techniques have a direct influence on the bioactivity and mechanical behavior of scaffolds. A detailed discussion of the bioactivity, in vitro and in vivo biocompatibility and biodegradation is presented as a function of materials and its processing techniques. Finally, an outlook for future research is also proposed. © 2017 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 105A: 3197-3223, 2017.
    Matched MeSH terms: Tissue Scaffolds/chemistry
  19. Sangkert S, Kamonmattayakul S, Chai WL, Meesane J
    J Biomed Mater Res A, 2017 Jun;105(6):1624-1636.
    PMID: 28000362 DOI: 10.1002/jbm.a.35983
    Maxillofacial bone defect is a critical problem for many patients. In severe cases, the patients need an operation using a biomaterial replacement. Therefore, to design performance biomaterials is a challenge for materials scientists and maxillofacial surgeons. In this research, porous silk fibroin scaffolds with mimicked microenvironment based on decellularized pulp and fibronectin were created as for bone regeneration. Silk fibroin scaffolds were fabricated by freeze-drying before modification with three different components: decellularized pulp, fibronectin, and decellularized pulp/fibronectin. The morphologies of the modified scaffolds were observed by scanning electron microscopy. Existence of the modifying components in the scaffolds was proved by the increase in weights and from the pore size measurements of the scaffolds. The modified scaffolds were seeded with MG-63 osteoblasts and cultured. Testing of the biofunctionalities included cell viability, cell proliferation, calcium content, alkaline phosphatase activity (ALP), mineralization and histological analysis. The results demonstrated that the modifying components organized themselves into aggregations of a globular structure. They were arranged themselves into clusters of aggregations with a fibril structure in the porous walls of the scaffolds. The results showed that modified scaffolds with a mimicked microenvironment of decellularized pulp/fibronectin were suitable for cell viability since the cells could attach and spread into most of the pores of the scaffold. Furthermore, the scaffolds could induce calcium synthesis, mineralization, and ALP activity. The results indicated that modified silk fibroin scaffolds with a mimicked microenvironment of decellularized pulp/fibronectin hold promise for use in tissue engineering in maxillofacial bone defects. © 2017 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 105A: 1624-1636, 2017.
    Matched MeSH terms: Tissue Scaffolds/chemistry*
  20. Heng BC, Gong T, Wang S, Lim LW, Wu W, Zhang C
    J Endod, 2017 Mar;43(3):409-416.
    PMID: 28231979 DOI: 10.1016/j.joen.2016.10.033
    INTRODUCTION: Dental follicle stem cells (DFSCs) possess neurogenic potential because they originate from the embryonic neural crest. This study investigated whether neural differentiation of DFSCs can be enhanced by culture on decellularized matrix substrata (NSC-DECM) derived from neurogenesis of human embryonic stem cells (hESCs).

    METHODS: The hESCs were differentiated into neural stem cells (NSCs), and NSC-DECM was extracted from confluent monolayers of NSCs through treatment with deionized water. DFSCs seeded on NSC-DECM, Geltrex, and tissue culture polystyrene (TCPS) were subjected to neural induction during a period of 21 days. Expression of early/intermediate (Musashi1, PAX6, NSE, and βIII-tubulin) and mature/late (NGN2, NeuN, NFM, and MASH1) neural markers by DFSCs was analyzed at the 7-, 14-, and 21-day time points with quantitative real-time polymerase chain reaction. Immunocytochemistry for detection of βIII-tubulin, PAX6, and NGN2 expression by DFSCs on day 7 of neural induction was also carried out.

    RESULTS: Quantitative RT-PCR showed that expression of PAX6, Musashi1, βIII-tubulin, NSE, NGN2, and NFM by DFSCs was enhanced on NSC-DECM versus either the Geltrex or TCPS groups. Immunocytochemistry showed that DFSCs in the NSC-DECM group displayed more intense staining for βIII-tubulin, PAX6, and NGN2 expression, together with more neurite outgrowths and elongated morphology, as compared with either Geltrex or TCPS.

    CONCLUSIONS: DECM derived from neurogenesis of hESCs can enhance the neurogenic potential of DFSCs.

    Matched MeSH terms: Tissue Scaffolds
Filters
Contact Us

Please provide feedback to Administrator (afdal@afpm.org.my)

External Links