Displaying publications 41 - 60 of 77 in total

Abstract:
Sort:
  1. Kouhi M, Jayarama Reddy V, Ramakrishna S
    Appl Biochem Biotechnol, 2019 Jun;188(2):357-368.
    PMID: 30456599 DOI: 10.1007/s12010-018-2922-0
    Bioceramic nanoparticles with high specific surface area often tend to agglomerate in the polymer matrix, which results in undesirable mechanical properties of the composites and poor cell spreading and attachment. In the present work, bredigite (BR) nanoparticles were modified with an organosilane coupling agent, 3-glycidoxypropyltrimethoxysilane (GPTMS), to enhance its dispersibility in the polymer matrix. The polyhydroxybutyrate-co-hydroxyvaletare (PHBV) nanofibrous scaffolds containing either bredigite or GPTMS-modified bredigite (G-BR) nanoparticles were fabricated using electrospinning technique and characterized using scanning electron microscopy, transmission electron microscopy, and tensile strength. Results demonstrated that modification of bredigite was effective in enhancing nanoparticle dispersion in the PHBV matrix. PHBV/G-BR scaffold showed improved mechanical properties compared to PHBV and PHBV/BR, especially at the higher concentration of nanoparticles. In vitro bioactivity assay performed in the simulated body fluid (SBF) indicated that composite PHBV scaffolds were able to induce the formation of apatite deposits after incubation in SBF. From the results of in vitro biological assay, it is concluded that the synergetic effect of BR and GPTMS provided an enhanced hFob cells attachment and proliferation. The developed PHBV/G-BR nanofibrous scaffolds may be considered for application in bone tissue engineering.
    Matched MeSH terms: Tissue Scaffolds/chemistry*
  2. Khan MUA, Haider S, Shah SA, Razak SIA, Hassan SA, Kadir MRA, et al.
    Int J Biol Macromol, 2020 May 15;151:584-594.
    PMID: 32081758 DOI: 10.1016/j.ijbiomac.2020.02.142
    Arabinoxylan (AX) is a natural biological macromolecule with several potential biomedical applications. In this research, AX, nano-hydroxyapatite (n-HAp) and titanium dioxide (TiO2) based polymeric nanocomposite scaffolds were fabricated by the freeze-drying method. The physicochemical characterizations of these polymeric nanocomposite scaffolds were performed for surface morphology, porosity, swelling, biodegradability, mechanical, and biological properties. The scaffolds exhibited good porosity and rough surface morphology, which were efficiently controlled by TiO2 concentrations. MC3T3-E1 cells were employed to conduct the biocompatibility of these scaffolds. Scaffolds showed unique biocompatibility in vitro and was favorable for cell attachment and growth. PNS3 proved more biocompatible, showed interconnected porosity and substantial mechanical strength compared to PNS1, PNS2 and PNS4. Furthermore, it has also showed more affinity to cells and cell growth. The results illustrated that the bioactive nanocomposite scaffold has the potential to find applications in the tissue engineering field.
    Matched MeSH terms: Tissue Scaffolds/chemistry*
  3. Khan MUA, Razak SIA, Rehman S, Hasan A, Qureshi S, Stojanović GM
    Int J Biol Macromol, 2022 Dec 01;222(Pt A):462-472.
    PMID: 36155784 DOI: 10.1016/j.ijbiomac.2022.09.153
    Globally, people suffering from bone disorders are steadily increasing and bone tissue engineering is an advanced approach to treating fractured and defected bone tissues. In this study, we have prepared polymeric nanocomposite by free-radical polymerization from sodium alginate, hydroxyapatite, and silica with different GO amounts. The porous scaffolds were fabricated using the freeze drying technique. The structural, morphological, mechanical, and wetting investigation was conducted by Fourier-transform infrared spectroscopy, X-ray diffraction, scanning electron microscope, universal tensile machine, and water contact angle characterization techniques. The swelling, biodegradation, and water retention were also studied. The biological studies were performed (cell viability, cell adherence, proliferation, and mineralization) against osteoblast cell lines. Scaffolds have exhibited different pore morphology SAG-1 (pore size = 414.61 ± 56 μm and porosity = 81.45 ± 2.17 %) and SAG-4 (pore size = 195.97 ± 82 μm and porosity = 53.82 ± 2.45 %). They have different mechanical behavior as SAG-1 has the least compression strength and compression modulus 2.14 ± 2.35 and 16.51 ± 1.27 MPa. However, SAG-4 has maximum compression strength and compression modulus 13.67 ± 2.63 and 96.16 ± 1.97 MPa with wetting behavior 80.70° and 58.70°, respectively. Similarly, SAG-1 exhibited the least and SAG-4 presented maximum apatite mineral formation, cell adherence, cell viability, and cell proliferation against mouse pre-osteoblast cell lines. The increased GO amount provides different multifunctional materials with different characteristics. Hence, the fabricated scaffolds could be potential scaffold materials to treat and regenerate fracture bone tissues in bone tissue engineering.
    Matched MeSH terms: Tissue Scaffolds/chemistry
  4. Kamarul T, Krishnamurithy G, Salih ND, Ibrahim NS, Raghavendran HR, Suhaeb AR, et al.
    ScientificWorldJournal, 2014;2014:905103.
    PMID: 25298970 DOI: 10.1155/2014/905103
    The in vivo biocompatibility and toxicity of PVA/NOCC scaffold were tested by comparing them with those of a biocompatible inert material HAM in a rat model. On Day 5, changes in the blood parameters of the PVA/NOCC-implanted rats were significantly higher than those of the control. The levels of potassium, creatinine, total protein, A/G, hemoglobulin, erythrocytes, WBC, and platelets were not significantly altered in the HAM-implanted rats, when compared with those in the control. On Day 10, an increase in potassium, urea, and GGT levels and a decrease in ALP, platelet, and eosinophil levels were noted in the PVA/NOCC-implanted rats, when compared with control. These changes were almost similar to those noted in the HAM-implanted rats, except for the unaltered potassium and increased neutrophil levels. On Day 15, the total protein, A/G, lymphocyte, monocyte, and eosinophil levels remained unaltered in the PVA/NOCC-implanted rats, whereas urea, A/G, WBC, lymphocyte, and monocyte levels remained unchanged in the HAM-implanted rats. Histology and immunohistochemistry analyses revealed inflammatory infiltration in the PVA/NOCC-implanted rats, but not in the HAM-implanted rats. Although a low toxic tissue response was observed in the PVA/NOCC-implanted rats, further studies are necessary to justify the use of this material in tissue engineering applications.
    Matched MeSH terms: Tissue Scaffolds/chemistry*
  5. Jithendra P, Mohamed JMM, Annamalai D, Al-Serwi RH, Ibrahim AM, El-Sherbiny M, et al.
    Int J Biol Macromol, 2023 Sep 01;248:125948.
    PMID: 37482169 DOI: 10.1016/j.ijbiomac.2023.125948
    The chondrogenic efficacy of aloe vera blended collagen-chitosan (COL-CS-AV) porous scaffold was investigated using articular chondrocytes in a standard condition. Cytocompatibility was analyzed using fluorescent dyes (calcein AM/ethidium bromide) and the viable cells were quantified by MTT assay. Glycosaminoglycan (GAG) content of ECM was estimated by using 1, 9-Dimethyl methylene Blue (DMMB). The total RNA content was quantified and the cartilage specific genes (col2a1, Acan) were amplified by reverse transcription-PCR from the cell lysate of the scaffolds. Histological examination was made using Haematoxylin and Eosin (H&E), safranin-O, masson's trichrome, alcian blue, and alizarin red to stain the specific component of ECM secreted on the construct. The cartilage specific collagen type II was estimated by immunohistochemistry using monoclonal type II collagen antibody. The results of these studies proved that COL-CS-AV scaffold has more chondrogenic efficacy than COL-CS, thus the aloe vera blend COL-CS-AV scaffold might be used as suitable candidate for cartilage tissue engineering.
    Matched MeSH terms: Tissue Scaffolds/chemistry
  6. Jiang H, Mani MP, Jaganathan SK
    Int J Nanomedicine, 2019;14:8149-8159.
    PMID: 31632024 DOI: 10.2147/IJN.S214646
    Introduction: Recently several new approaches were emerging in bone tissue engineering to develop a substitute for remodelling the damaged tissue. In order to resemble the native extracellular matrix (ECM) of the human tissue, the bone scaffolds must possess necessary requirements like large surface area, interconnected pores and sufficient mechanical strength.

    Materials and methods: A novel bone scaffold has been developed using polyurethane (PE) added with wintergreen (WG) and titanium dioxide (TiO2). The developed nanocomposites were characterized through field emission scanning electron microscopy (FESEM), Fourier transform and infrared spectroscopy (FTIR), X-ray diffraction (XRD), contact angle measurement, thermogravimetric analysis (TGA), atomic force microscopy (AFM) and tensile testing. Furthermore, anticoagulant assays, cell viability analysis and calcium deposition were used to investigate the biological properties of the prepared hybrid nanocomposites.

    Results: FESEM depicted the reduced fibre diameter for the electrospun PE/WG and PE/WG/TiO2 than the pristine PE. The addition of WG and TiO2 resulted in the alteration in peak intensity of PE as revealed in the FTIR. Wettability measurements showed the PE/WG showed decreased wettability and the PE/WG/TiO2 exhibited improved wettability than the pristine PE. TGA measurements showed the improved thermal behaviour for the PE with the addition of WG and TiO2. Surface analysis indicated that the composite has a smoother surface rather than the pristine PE. Further, the incorporation of WG and TiO2 improved the anticoagulant nature of the pristine PE. In vitro cytotoxicity assay has been performed using fibroblast cells which revealed that the electrospun composites showed good cell attachment and proliferation after 5 days. Moreover, the bone apatite formation study revealed the enhanced deposition of calcium content in the fabricated composites than the pristine PE.

    Conclusion: Fabricated nanocomposites rendered improved physico-chemical properties, biocompatibility and calcium deposition which are conducive for bone tissue engineering.

    Matched MeSH terms: Tissue Scaffolds/chemistry*
  7. Hu Y, Ran J, Zheng Z, Jin Z, Chen X, Yin Z, et al.
    Acta Biomater, 2018 04 15;71:168-183.
    PMID: 29524675 DOI: 10.1016/j.actbio.2018.02.019
    Anterior cruciate ligament (ACL) is one of the most difficult tissues to heal once injured. Ligament regeneration and tendon-bone junction healing are two major goals of ACL reconstruction. This study aimed to investigate the synergistic therapeutic effects of Stromal cell-derived factor 1 (SDF-1)-releasing collagen-silk (CSF) scaffold combined with intra-articular injection of ligament-derived stem/progenitor cells (LSPCs) for ACL regeneration and the amelioration in the long-term complication of osteoarthritis (OA). The stem cell recruitment ability of CSF scaffold and the multipotency, particularly the tendon forming ability of LSPCs from rabbits were characterized in vitro, while the synergistic effect of the CSF scaffold and LSPCs for ACL regeneration and OA amelioration were investigated in vivo at 1, 3, and 6 months with a rabbit ACL reconstruction model. The CSF scaffold was used as a substitute for the ACL, and LSPCs were injected into the joint cavity after 7 days of the ACL reconstruction. CSF scaffold displayed a controlled release pattern for the encapsulated protein for up to 7 days with an increased stiffness in the mechanical property. LSPCs, which exhibited highly I Collagen and CXCR4 expression, were attracted by SDF-1 and successfully relocated into the CSF scaffold at 1 month in vivo. At 3 and 6 months post-treatment, the CSF scaffold combined with LSPCs (CSFL group) enhanced the regeneration of ACL tissue, and promoted bone tunnel healing. Furthermore, the OA progression was impeded efficiently. Our findings here provided a new strategy that using stem cell recruiting CSF scaffold with tissue-specific stem cells, could be a promising solution for ACL regeneration.

    STATEMENT OF SIGNIFICANCE: In this study, we developed a silk scaffold with increased stiffness and SDF-1 controlled release capacity for ligament repair. This advanced scaffold transplantation combined with intra-articular injection of LSPCs (which was isolated from rabbit ligament for the first time in this study) promoted the regeneration of both the tendinous and bone tunnel portion of ACL. This therapeutic strategy also ameliorated cartilage degeneration and reduced the severity of arthrofibrosis. Hence, combining LSPCs injection with SDF-1-releasing silk scaffold is demonstrated as a therapeutic strategy for ACL regeneration and OA treatment in the clinic.

    Matched MeSH terms: Tissue Scaffolds/chemistry*
  8. Hoque ME, Chuan YL, Pashby I
    Biopolymers, 2012 Feb;97(2):83-93.
    PMID: 21830198 DOI: 10.1002/bip.21701
    Advances in scaffold design and fabrication technology have brought the tissue engineering field stepping into a new era. Conventional techniques used to develop scaffolds inherit limitations, such as lack of control over the pore morphology and architecture as well as reproducibility. Rapid prototyping (RP) technology, a layer-by-layer additive approach offers a unique opportunity to build complex 3D architectures overcoming those limitations that could ultimately be tailored to cater for patient-specific applications. Using RP methods, researchers have been able to customize scaffolds to mimic the biomechanical properties (in terms of structural integrity, strength, and microenvironment) of the organ or tissue to be repaired/replaced quite closely. This article provides intensive description on various extrusion based scaffold fabrication techniques and review their potential utility for TE applications. The extrusion-based technique extrudes the molten polymer as a thin filament through a nozzle onto a platform layer-by-layer and thus building 3D scaffold. The technique allows full control over pore architecture and dimension in the x- and y- planes. However, the pore height in z-direction is predetermined by the extruding nozzle diameter rather than the technique itself. This review attempts to assess the current state and future prospects of this technology.
    Matched MeSH terms: Tissue Scaffolds/chemistry*
  9. Hoque ME, San WY, Wei F, Li S, Huang MH, Vert M, et al.
    Tissue Eng Part A, 2009 Oct;15(10):3013-24.
    PMID: 19331580 DOI: 10.1089/ten.TEA.2008.0355
    Synthetic polymers have attracted much attention in tissue engineering due to their ability to modulate biomechanical properties. This study investigated the feasibility of processing poly(epsilon-caprolactone) (PCL) homopolymer, PCL-poly(ethylene glycol) (PEG) diblock, and PCL-PEG-PCL triblock copolymers into three-dimensional porous scaffolds. Properties of the various polymers were investigated by dynamic thermal analysis. The scaffolds were manufactured using the desktop robot-based rapid prototyping technique. Gross morphology and internal three-dimensional structure of scaffolds were identified by scanning electron microscopy and micro-computed tomography, which showed excellent fusion at the filament junctions, high uniformity, and complete interconnectivity of pore networks. The influences of process parameters on scaffolds' morphological and mechanical characteristics were studied. Data confirmed that the process parameters directly influenced the pore size, porosity, and, consequently, the mechanical properties of the scaffolds. The in vitro cell culture study was performed to investigate the influence of polymer nature and scaffold architecture on the adhesion of the cells onto the scaffolds using rabbit smooth muscle cells. Light, scanning electron, and confocal laser microscopy showed cell adhesion, proliferation, and extracellular matrix formation on the surface as well as inside the structure of both scaffold groups. The completely interconnected and highly regular honeycomb-like pore morphology supported bridging of the pores via cell-to-cell contact as well as production of extracellular matrix at later time points. The results indicated that the incorporation of hydrophilic PEG into hydrophobic PCL enhanced the overall hydrophilicity and cell culture performance of PCL-PEG copolymer. However, the scaffold architecture did not significantly influence the cell culture performance in this study.
    Matched MeSH terms: Tissue Scaffolds/chemistry*
  10. Hazwani A, Sha'Ban M, Azhim A
    Organogenesis, 2019;15(4):120-136.
    PMID: 31495272 DOI: 10.1080/15476278.2019.1656997
    Extracellular matrix (ECM) based bioscaffolds prepared by decellularization has increasingly emerged in tissue engineering application because it has structural, biochemical, and biomechanical cues that have dramatic effects upon cell behaviors. Therefore, we developed a closed sonication decellularization system to prepare ideal bioscaffolds with minimal adverse effects on the ECM. The decellularization was achieved at 170 kHz of ultrasound frequency in 0.1% and 2% Sodium Dodecyl Sulphate (SDS) solution for 10 hours. The immersion treatment as control was performed to compare the decellularization efficiency with our system. Cell removal and ECM structure were determined by histological staining and biochemical assay. Biomechanical properties were investigated by the indentation testing to test the stiffness, a residual force and compression of bioscaffolds. Additionally, in vivo implantation was performed in rat to investigate host tissue response. Compared to native tissues, histological staining and biochemical assay confirm the absence of cellularity with preservation of ECM structure. Moreover, sonication treatment has not affected the stiffness [N/mm] and a residual force [N] of the aortic scaffolds except for compression [%] which 2% SDS significantly decreased compared to native tissues showing higher SDS has a detrimental effect on ECM structure. Finally, minimal inflammatory response was observed after 1 and 5 weeks of implantation. This study reported that the novelty of our developed closed sonication system to prepare ideal bioscaffolds for tissue engineering applications.
    Matched MeSH terms: Tissue Scaffolds/chemistry*
  11. Haniffa MACM, Munawar K, Chee CY, Pramanik S, Halilu A, Illias HA, et al.
    Carbohydr Polym, 2021 Sep 01;267:118136.
    PMID: 34119125 DOI: 10.1016/j.carbpol.2021.118136
    Cellulose and its forms are widely used in biomedical applications due to their biocompatibility, biodegradability and lack of cytotoxicity. It provides ample opportunities for the functionalization of supported magnetic nanohybrids (CSMNs). Because of the abundance of surface hydroxyl groups, they are surface tunable in either homogeneous or heterogeneous solvents and thus act as a substrate or template for the CSMNs' development. The present review emphasizes on the synthesis of various CSMNs, their physicomagnetic properties, and potential applications such as stimuli-responsive drug delivery systems, MRI, enzyme encapsulation, nucleic acid extraction, wound healing and tissue engineering. The impact of CSMNs on cytotoxicity, magnetic hyperthermia, and folate-conjugates is highlighted in particular, based on their structures, cell viability, and stability. Finally, the review also discussed the challenges and prospects of CSMNs' development. This review is expected to provide CSMNs' development roadmap in the context of 21st-century demands for biomedical therapeutics.
    Matched MeSH terms: Tissue Scaffolds/chemistry
  12. Gorain B, Choudhury H, Pandey M, Kesharwani P, Abeer MM, Tekade RK, et al.
    Biomed Pharmacother, 2018 Aug;104:496-508.
    PMID: 29800914 DOI: 10.1016/j.biopha.2018.05.066
    Myocardial infarction (cardiac tissue death) is among the most prevalent causes of death among the cardiac patients due to the inability of self-repair in cardiac tissues. Myocardial tissue engineering is regarded as one of the most realistic strategies for repairing damaged cardiac tissue. However, hindrance in transduction of electric signals across the cardiomyocytes due to insulating properties of polymeric materials worsens the clinical viability of myocardial tissue engineering. Aligned and conductive scaffolds based on Carbon nanotubes (CNT) have gained remarkable recognition due to their exceptional attributes which provide synthetic but viable microenvironment for regeneration of engineered cardiomyocytes. This review presents an overview and critical analysis of pharmaceutical implications and therapeutic feasibility of CNT based scaffolds in improving the cardiac tissue regeneration and functionality. The expository analysis of the available evidence revealed that inclusion of single- or multi-walled CNT into fibrous, polymeric, and elastomeric scaffolds results in significant improvement in electrical stimulation and signal transduction through cardiomyocytes. Moreover, incorporation of CNT in engineering scaffolds showed a greater potential of augmenting cardiomyocyte proliferation, differentiation, and maturation and has improved synchronous beating of cardiomyocytes. Despite promising ability of CNT in promoting functionality of cardiomyocytes, their presence in scaffolds resulted in substantial improvement in mechanical properties and structural integrity. Conclusively, this review provides new insight into the remarkable potential of CNT aligned scaffolds in improving the functionality of engineered cardiac tissue and signifies their feasibility in cardiac tissue regenerative medicines and stem cell therapy.
    Matched MeSH terms: Tissue Scaffolds/chemistry*
  13. 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*
  14. Gnaneshwar PV, Sudakaran SV, Abisegapriyan S, Sherine J, Ramakrishna S, Rahim MHA, et al.
    Mater Sci Eng C Mater Biol Appl, 2019 Mar;96:337-346.
    PMID: 30606541 DOI: 10.1016/j.msec.2018.11.033
    Far-flung evolution in tissue engineering enabled the development of bioactive and biodegradable materials to generate biocomposite nanofibrous scaffolds for bone repair and replacement therapies. Polymeric bioactive nanofibers are to biomimic the native extracellular matrix (ECM), delivering tremendous regenerative potentials for drug delivery and tissue engineering applications. It's been known from few decades that Zinc oxide (ZnO) nanoparticles are enhancing bone growth and providing proliferation of osteoblasts when incorporated with hydroxyapatite (HAp). We attempted to investigate the interaction between the human foetal osteoblasts (hFOB) with ZnO doped HAp incorporated biocomposite poly(L-lactic acid)-co-poly(ε-caprolactone) and silk fibroin (PLACL/SF) nanofibrous scaffolds for osteoblasts mineralization in bone tissue regeneration. The present study, we doped ZnO with HAp (ZnO(HAp) using the sol-gel ethanol condensation technique. The properties of PLACL/SF/ZnO(HAp) biocomposite nanofibrous scaffolds enhanced with doped and blended ZnO/HAp were characterized using Scanning electron microscopy (SEM), Fourier transform infrared spectroscopy (FTIR), Contact angle and Tensile studies to determine the morphology, functionality, wettability and stability. The in vitro study results showed that the addition of ZnO and HAp enhances the secretion of bone mineral matrix (98%) with smaller fiber diameter (139.4 ± 27 nm) due to the presence of silk fibroin showing potential tensile properties (322.4%), and increased the proliferation of osteoblasts for bone tissue regeneration.
    Matched MeSH terms: Tissue Scaffolds/chemistry*
  15. Geetha Bai R, Muthoosamy K, Manickam S, Hilal-Alnaqbi A
    Int J Nanomedicine, 2019;14:5753-5783.
    PMID: 31413573 DOI: 10.2147/IJN.S192779
    Tissue engineering embraces the potential of recreating and replacing defective body parts by advancements in the medical field. Being a biocompatible nanomaterial with outstanding physical, chemical, optical, and biological properties, graphene-based materials were successfully employed in creating the perfect scaffold for a range of organs, starting from the skin through to the brain. Investigations on 2D and 3D tissue culture scaffolds incorporated with graphene or its derivatives have revealed the capability of this carbon material in mimicking in vivo environment. The porous morphology, great surface area, selective permeability of gases, excellent mechanical strength, good thermal and electrical conductivity, good optical properties, and biodegradability enable graphene materials to be the best component for scaffold engineering. Along with the apt microenvironment, this material was found to be efficient in differentiating stem cells into specific cell types. Furthermore, the scope of graphene nanomaterials in liver tissue engineering as a promising biomaterial is also discussed. This review critically looks into the unlimited potential of graphene-based nanomaterials in future tissue engineering and regenerative therapy.
    Matched MeSH terms: Tissue Scaffolds/chemistry*
  16. Fauzi MB, Lokanathan Y, Aminuddin BS, Ruszymah BHI, Chowdhury SR
    Mater Sci Eng C Mater Biol Appl, 2016 Nov 01;68:163-171.
    PMID: 27524008 DOI: 10.1016/j.msec.2016.05.109
    Collagen is the most abundant extracellular matrix (ECM) protein in the human body, thus widely used in tissue engineering and subsequent clinical applications. This study aimed to extract collagen from ovine (Ovis aries) Achilles tendon (OTC), and to evaluate its physicochemical properties and its potential to fabricate thin film with collagen fibrils in a random or aligned orientation. Acid-solubilized protein was extracted from ovine Achilles tendon using 0.35M acetic acid, and 80% of extracted protein was measured as collagen. SDS-PAGE and mass spectrometry analysis revealed the presence of alpha 1 and alpha 2 chain of collagen type I (col I). Further analysis with Fourier transform infrared spectrometry (FTIR), X-ray diffraction (XRD) and energy dispersive X-ray spectroscopy (EDS) confirms the presence of triple helix structure of col I, similar to commercially available rat tail col I. Drying the OTC solution at 37°C resulted in formation of a thin film with randomly orientated collagen fibrils (random collagen film; RCF). Introduction of unidirectional mechanical intervention using a platform rocker prior to drying facilitated the fabrication of a film with aligned orientation of collagen fibril (aligned collagen film; ACF). It was shown that both RCF and ACF significantly enhanced human dermal fibroblast (HDF) attachment and proliferation than that on plastic surface. Moreover, cells were distributed randomly on RCF, but aligned with the direction of mechanical intervention on ACF. In conclusion, ovine tendon could be an alternative source of col I to fabricate scaffold for tissue engineering applications.
    Matched MeSH terms: Tissue Scaffolds/chemistry*
  17. Fallahiarezoudar E, Ahmadipourroudposht M, Idris A, Mohd Yusof N
    Mater Sci Eng C Mater Biol Appl, 2015 Mar;48:556-65.
    PMID: 25579957 DOI: 10.1016/j.msec.2014.12.016
    The four heart valves represented in the mammalian hearts are responsible for maintaining unidirectional, non-hinder blood flow. The heart valve leaflets synchronically open and close approximately 4 million times a year and more than 3 billion times during the life. Valvular heart dysfunction is a significant cause of morbidity and mortality around the world. When one of the valves malfunctions, the medical choice is may be to replace the original valves with an artificial one. Currently, the mechanical and biological artificial valves are clinically used with some drawbacks. Tissue engineering heart valve concept represents a new technique to enhance the current model. In tissue engineering method, a three-dimensional scaffold is fabricated as the template for neo-tissue development. Appropriate cells are seeded to the matrix in vitro. Various approaches have been investigated either in scaffold biomaterials and fabrication techniques or cell source and cultivation methods. The available results of ongoing experiments indicate a promising future in this area (particularly in combination of bone marrow stem cells with synthetic scaffold), which can eliminate the need for lifelong anti-coagulation medication, durability and reoperation problems.
    Matched MeSH terms: Tissue Scaffolds/chemistry*
  18. Ezhilarasu H, Sadiq A, Ratheesh G, Sridhar S, Ramakrishna S, Ab Rahim MH, et al.
    Nanomedicine (Lond), 2019 01;14(2):201-214.
    PMID: 30526272 DOI: 10.2217/nnm-2018-0271
    AIM: Atherosclerosis is a common cardiovascular disease causing medical problems globally leading to coronary artery bypass surgery. The present study is to fabricate core/shell nanofibers to encapsulate VEGF for the differentiation of mesenchymal stem cells (MSCs) into smooth muscle cells to develop vascular grafts.

    MATERIALS & METHODS: The fabricated core/shell nanofibers contained polycaprolactone/gelatin as the shell, and silk fibroin/VEGF as the core materials.

    RESULTS: The results observed that the core/shell nanofibers interact to differentiate MSCs into smooth muscle cells by the expression of vascular smooth muscle cell (VSMC) contractile proteins α-actinin, myosin and F-actin.

    CONCLUSION: The functionalized polycaprolactone/gelatin/silk fibroin/VEGF (250 ng) core/shell nanofibers were fabricated for the controlled release of VEGF in a persistent manner for the differentiation of MSCs into smooth muscle cells for vascular tissue engineering.

    Matched MeSH terms: Tissue Scaffolds/chemistry
  19. Ebrahimi S, Hanim YU, Sipaut CS, Jan NBA, Arshad SE, How SE
    Int J Mol Sci, 2021 Sep 06;22(17).
    PMID: 34502544 DOI: 10.3390/ijms22179637
    Recently, composite scaffolding has found many applications in hard tissue engineering due to a number of desirable features. In this present study, hydroxyapatite/bioglass (HAp/BG) nanocomposite scaffolds were prepared in different ratios using a hydrothermal approach. The aim of this research was to evaluate the adhesion, growth, viability, and osteoblast differentiation behavior of human Wharton's-jelly-derived mesenchymal stem cells (hWJMSCs) on HAp/BG in vitro as a scaffold for application in bone tissue engineering. Particle size and morphology were investigated by TEM and bioactivity was assessed and proven using SEM analysis with hWJMSCs in contact with the HAp/BG nanocomposite. Viability was evaluated using PrestoBlueTM assay and early osteoblast differentiation and mineralization behaviors were investigated by ALP activity and EDX analysis simultaneously. TEM results showed that the prepared HAp/BG nanocomposite had dimensions of less than 40 nm. The morphology of hWJMSCs showed a fibroblast-like shape, with a clear filopodia structure. The viability of hWJMSCs was highest for the HAp/BG nanocomposite with a 70:30 ratio of HAp to BG (HAp70/BG30). The in vitro biological results confirmed that HAp/BG composite was not cytotoxic. It was also observed that the biological performance of HAp70/BG30 was higher than HAp scaffold alone. In summary, HAp/BG scaffold combined with mesenchymal stem cells showed significant potential for bone repair applications in tissue engineering.
    Matched MeSH terms: Tissue Scaffolds/chemistry
  20. Dhand C, Balakrishnan Y, Ong ST, Dwivedi N, Venugopal JR, Harini S, et al.
    Int J Nanomedicine, 2018;13:4473-4492.
    PMID: 30122921 DOI: 10.2147/IJN.S159770
    Introduction: In search for cross-linkers with multifunctional characteristics, the present work investigated the utility of quaternary ammonium organosilane (QOS) as a potential cross-linker for electrospun collagen nanofibers. We hypothesized that the quaternary ammonium ions improve the electrospinnability by reducing the surface tension and confer antimicrobial properties, while the formation of siloxane after alkaline hydrolysis could cross-link collagen and stimulate cell proliferation.

    Materials and methods: QOS collagen nanofibers were electrospun by incorporating various concentrations of QOS (0.1%-10% w/w) and were cross-linked in situ after exposure to ammonium carbonate. The QOS cross-linked scaffolds were characterized and their biological properties were evaluated in terms of their biocompatibility, cellular adhesion and metabolic activity for primary human dermal fibroblasts and human fetal osteoblasts.

    Results and discussion: The study revealed that 1) QOS cross-linking increased the flexibility of otherwise rigid collagen nanofibers and improved the thermal stability; 2) QOS cross-linked mats displayed potent antibacterial activity and 3) the biocompatibility of the composite mats depended on the amount of QOS present in dope solution - at low QOS concentrations (0.1% w/w), the mats promoted mammalian cell proliferation and growth, whereas at higher QOS concentrations, cytotoxic effect was observed.

    Conclusion: This study demonstrates that QOS cross-linked mats possess anti-infective properties and confer niches for cellular growth and proliferation, thus offering a useful approach, which is important for hard and soft tissue engineering and regenerative medicine.

    Matched MeSH terms: Tissue Scaffolds/chemistry*
Filters
Contact Us

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

External Links